EP1169348B1 - Purposeful movement of human migratory cells away from an agent source - Google Patents

Purposeful movement of human migratory cells away from an agent source Download PDF

Info

Publication number
EP1169348B1
EP1169348B1 EP00922067.4A EP00922067A EP1169348B1 EP 1169348 B1 EP1169348 B1 EP 1169348B1 EP 00922067 A EP00922067 A EP 00922067A EP 1169348 B1 EP1169348 B1 EP 1169348B1
Authority
EP
European Patent Office
Prior art keywords
cells
cell
fugetactic
agent
sdf
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
EP00922067.4A
Other languages
German (de)
French (fr)
Other versions
EP1169348A1 (en
Inventor
Mark C. Poznansky
Andrew D. Luster
David T. Scadden
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
General Hospital Corp
Original Assignee
General Hospital Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by General Hospital Corp filed Critical General Hospital Corp
Publication of EP1169348A1 publication Critical patent/EP1169348A1/en
Application granted granted Critical
Publication of EP1169348B1 publication Critical patent/EP1169348B1/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/502Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects
    • G01N33/5029Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects on cell motility
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/521Chemokines
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/81Protease inhibitors
    • C07K14/8107Endopeptidase (E.C. 3.4.21-99) inhibitors
    • C07K14/811Serine protease (E.C. 3.4.21) inhibitors
    • C07K14/8114Kunitz type inhibitors
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5011Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5047Cells of the immune system
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5047Cells of the immune system
    • G01N33/505Cells of the immune system involving T-cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5058Neurological cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5064Endothelial cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5082Supracellular entities, e.g. tissue, organisms
    • G01N33/5088Supracellular entities, e.g. tissue, organisms of vertebrates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • This invention relates to methods and compositions for modulating movement of eukaryotic cells with migratory capacity. More specifically, there is described methods and compositions for modulating movement of cells of hematopoietic, neural, epithelial, or mesenchymal origin, in a specific site in a subject. The foregoing are useful, inter alia, in the treatment of conditions characterized by a need to modulate migratory-cell movement associated with specific sites in a subject. Specific sites include sites of inflammation and modulation of migratory-cell movement is movement away from an agent source, or repulsion.
  • Coli chemotaxis has revealed that a chemical which attracts the bacteria at some concentrations and conditions may also act as a negative chemotactic chemical or chemorepellent at others (Tsang N et al., 1973; Repaske D and Adler J. 1981; Tisa LS and Adler J.,1995; Taylor BL and Johnson MS., 1998).
  • Chemotaxis and chemokinesis have been observed to occur in mammalian cells (McCutcheon MW, Wartman W and HM Dixon, 1934; Lotz M and H Harris; 1956; Boyden SV 1962) in response to the class of proteins, called chemokines (Ward SG and Westwick J ; 1998; Kim CH et al., 1998; Baggiolini M , 1998; Farber JM; 1997).
  • Chemokines induce cell locomotion by signaling through G-protein coupled receptors (Wells TN et al.,1998).
  • the membrane protein gene superfamily of G-protein coupled receptors has been characterized as having seven putative transmembrane domains. The domains are believed to represent transmembrane ⁇ -helices connected by extracellular or cytoplasmic loops.
  • G-protein coupled receptors include a wide range of biologically active receptors, such as hormone, viral, growth factor and neuroreceptors.
  • G-protein coupled receptors have been characterized as including these seven conserved hydrophobic stretches of about 20 to 30 amino acids, connecting at least eight divergent hydrophilic loops.
  • the G-protein family of coupled receptors includes dopamine receptors which bind to neuroleptic drugs used for treating psychotic and neurological disorders.
  • Other examples of members of this family include calcitonin, adrenergic, endothelin, cAMP, adenosine, muscarinic, acetylcholine, serotonin, histamine, thrombin, kinin, follicle stimulating hormone, opsins, endothelial differentiation gene-1 receptor, rhodopsins, odorant, cytomegalovirus receptors, etc.
  • G-protein coupled receptors have single conserved cysteine residues in each of the first two extracellular loops which form disulfide bonds that are believed to stabilize functional protein structure.
  • the 7 transmembrane regions are designated as TM1, TM2, TM3, TM4,TM5, TM6, and TM7.
  • TM3 has been implicated in signal transduction.
  • Phosphorylation and lipidation (palmitylation or farnesylation) of cysteine residues can influence signal transduction of some G-protein coupled receptors.
  • Most G-protein coupled receptors contain potential phosphorylation sites within the third cytoplasmic loop and/or the carboxy-terminus. For several G-protein coupled receptors, such as the ⁇ -adrenoreceptor, phosphorylation by protein kinase A and/or specific receptor kinases mediates receptor desensitization.
  • the ligand binding sites of G-protein coupled receptors are believed to comprise a hydrophilic socket formed by several G-protein coupled receptors transmembrane domains, which socket is surrounded by hydrophobic residues of the G-protein coupled receptors.
  • the hydrophilic side of each G-protein coupled receptor transmembrane helix is postulated to face inward and form the polar ligand binding site.
  • TM3 has been implicated in several G-protein coupled receptors as having a ligand binding site, such as including the TM3 aspartate residue.
  • TM5 serines, a TM6 asparagine and TM6 or TM7 phenylalanines or tyrosines are also implicated in ligand binding.
  • G-protein coupled receptors can be intracellularly coupled by heterotrimeric G-proteins to various intracellular enzymes, ion channels and transporters (see, Johnson et al., Endoc Rev, 1989, 10:317-331 ). Different G-protein a-subunits preferentially stimulate particular effectors to modulate various biological functions in a cell. Phosphorylation of cytoplasmic residues of G-protein coupled receptors have been identified as an important mechanism for the regulation of G-protein coupling of some G-protein coupled receptors. G-protein coupled receptors are found in numerous sites within a mammalian host.
  • Chemokine-induced cell chemotaxis and chemokinesis are thought to be mediated via a G ⁇ i -linked signal transduction pathway and can be blocked by pertussis toxin (PTX) (Luster AD, 1998; Baggiolini, 1998).
  • PTX pertussis toxin
  • mice engineered to be deficient in SDF-1 ⁇ or its receptor, CXCR-4 have abnormal development of hematopoietic tissues and B-cells manifesting a failure of fetal liver stem cells to migrate to bone marrow (Friedland JS, 1995; Tan J and Thestrup-Pedersen K, 1995; Corrigan CJ and Kay AB, 1996; Qing M, et al,1998; Ward SG et al. 1998).
  • a fugetactic agent in the manufacture of a medicament for treating inflammation, autoimmune disease, an abscess, a transplant, an implant, atherosclerosis, myocarditis, infertility, or a tumor, the medicament containing a fugetactic amount of said agent for repelling immune cells from a specific site in a subject when the medicament is locally administered to the specific site in the subject, wherein the fugetactic agent is an agent having migratory cell repellent activity on a cell in a fugetactic amount, and having chemoattractant activity on the same cell at lower concentrations, wherein the fugetactic agent is SDF-1 ⁇ or HIV-I III gp120.
  • a fugetactic agent in the manufacture of a medicament containing a fugetactic amount of said agent for repelling immune cells away from a specific site in a subject, wherein the fugetactic agent is SDF-1 ⁇ or HIV- IIIB gp120.
  • a medicament containing a fugetactic agent for use in treating inflammation, autoimmune disease, an abscess, a transplant, an implant, atherosclerosis, myocarditis, infertility, or a tumor wherein the medicament contains a fugetactic amount of said agent for repelling immune cells from a specific site in a subject when the medicament is locally administered to the specific site in the subject, wherein the fugetactic agent is an agent having migratory cell repellent activity on a cell in a fugetactic amount, and having chemoattractant activity on the same cell at lower concentrations, wherein the fugetactic agent is SDF-1 ⁇ or HIV-I IIIB gp120.
  • a fugetactic agent to coat a material surface of an implant wherein the fugetactic agent is present in amount effective to repel immune cells from the material surface, wherein the fugetactic agent is SDF-1 ⁇ or HIV-I IIIB gp120.
  • an implant suitable for implanting into a subject wherein the implant comprises a material surface coated with an amount of a fugetactic agent effective to repel immune calls from the material surface, wherein the fugetactic agent is SDF-1 ⁇ or HIV-I IIIB gp120.
  • the method involves locally administering to a specific site in a subject in need of such treatment a fugetactic agent in an amount effective to inhibit migration of immune cells to the specific site in a subject.
  • the specific site may be a site of inflammation.
  • the method may further comprise co-administering a non-fugetactic agent that inhibits migration of immune cells to the site of inflammation in the subject.
  • the non-fugetactic agent may include an anti-inflammatory agent and/or an immunosuppressant.
  • the subject may have an autoimmune disease.
  • the autoimmune disease may include rheumatoid arthritis, uveitis, insulin-dependent diabetes mellitus, hemolytic anemias, rheumatic fever, Crohn's disease, Guillain-Barre syndrome, psoriasis, thyroiditis, Graves' disease, myasthenia gravis, glomerulonephritis, autoimmune hepatitis, systemic lupus erythematosus.
  • the subject has multiple sclerosis, an abscess, a transplant, an implant, atherosclerosis, and/or myocarditis.
  • the fugetactic agent is HIV-I IIIB gp120 or SDF-1 ⁇ .
  • the fugetactic agent is SDF-1 ⁇ at a concentration higher than about 1 ⁇ g/ml or HIV-1 IIIB gp120 at a concentration higher than about 200ng/mL
  • the method involves locally-administering to a specific site in a subject in need of such treatment a fugetactic agent in an amount effective to promote migration of cells away from the specific site in the subject.
  • the specific site may be an inflammation site.
  • the specific site may be a germ cell-containing site.
  • the specific site may be an area surrounding a tumor, but not the immediate area surrounding the tumor.
  • the specific site may be a transplanted tissue and/or an implant.
  • Important fugetactic agents, cell-types, and so on, are as described above. In any of the foregoing description, preferred cells are immune cells.
  • a method for distinguishing mature T-cells from immature T-cells in vitro involves contacting SDF-1 or HIV iiib gp120 with a mixed population of mature and immature T-cells and measuring cell movement relative to the fugetactic agent, wherein movement of cells away from the fugetactic agent is indicative of mature T-cells.
  • the method involves contacting an agent suspected of being a fugetactic agent with a cell with migratory capacity, measuring movement of the cell with migratory capacity relative to the agent, and determining whether the movement of the cell with migratory capacity is movement away from the agent, wherein movement of the cell with migratory capacity away from the agent is indicative of the agent being a fugetactic agent.
  • the cell with migratory capacity may be a hematopoietic cell.
  • the cell with migratory capacity may be a neural cell.
  • the cell with migratory capacity may be an epithelial cell.
  • the cell with migratory capacity may be a mesenchymal cell.
  • the cell with migratory capacity may be an embryonic stem cell.
  • the cell with migratory capacity may be a germ cell.
  • the agent depicted of being a fugetactic agent may be known to be a chemoattractant at a defined concentration and a fugetactic agent at a concentration higher than the defined concentration.
  • Such fugetactic agents may include cytokines.
  • the agent suspected of being a fugetactic agent may be an agent present in a biological fluid.
  • the biological fluid may include synovial fluid, cerebral spinal fluid, fellopian tube fluid, seminal fluid, ocular fluid, pericardial fluid, pleural fluid, inflammatory exudate and ascitic fluid.
  • the agent suspected of being a fugetactic agent may be an agent present in a tumor cell culture supernatant, tumor cell eluate and/or tumor cell lysate.
  • the tumor cells may be HepG2 cells and the cell with migratory capacity may be a hematopoietic cell.
  • the agent suspected of being a fugetactic agent may be an expression product of a cDNA library.
  • the method involves coating a material surface with an amount of a fugetactic agent effective to repel immune cells from the material surface.
  • the material surface may be part of an implant.
  • the material comprising the implant may be synthetic material or organic tissue material.
  • Important fugetactic agents, cell-types, and so on, are as described above.
  • the method involves forming a mixture comprising a cell with migratory capacity, a fugetactic agent and a candidate anti-fugetactic agent incubating the mixture under conditions which permit binding of the candidate anti-fugetactic agent to the fugetactic agent, determining the level of a test fugetactic activity of the fugetactic agent and the candidate anti-fugetactic agent on the cells with migratory capacity, and comparing the level of the test fugetactic activity to a control level of fugetactic activity determined in the absence of the candidate anti-fugetactic agent.
  • Reduction in the test fugetactic activity level relative to the control level of fugetactic activity may indicate that the candidate anti-fugetactic agent is a lead compound for an anti-fugetactic agent which inhibits the fugetactic activity of the fugetactic agent.
  • An increase in the test fugetactic activity level relative to control level of fugetactic activity may indicate that the candidate anti-fugetactic agent is a lead compound for an anti-fugetactic agent which increases the fugetactic activity of the fugetactic agent.
  • the fugetactic agent may be SDF-1 ⁇ at a concentration higher than about 1 ⁇ g/ml or HIV-1 IIIB gp120 at a concentration higher than about 200ng/ml.
  • the cell with migratory capacity may be a hematopoietic cell, a neural cell, an epithelial cell, a mesenchymal cell, an embryonic stem cell, a cell involved in angiogenesis (blood vessel formation), or a germ cell.
  • the method involves locally administering to a specific site in a subject in need of such treatment an anti-fugetactic agent in an amount effective to inhibit immune cell-specific fugetactic activity at the specific site in the subject.
  • the specific site may be a site of a pathogenic infection.
  • the specific site may be a germ cell-containing site.
  • the specific site may be an area immediately surrounding a tumor.
  • the anti-fugetactic agent described herein may be a cytokine binding agent such as an anti-cytokine antibody or a cytokine agonist.
  • the cytokine may be SDF-1 ⁇ .
  • the method involves locally administering to a tumor site in a subject in need of such treatment an anti-fugetactic agent in an amount effective to inhibit metastasis of tumor cells from the tumor site in the subject.
  • the anti-fugetactic agent may be as described above.
  • the method involves locally administering to an area surrounding a tumor site in a subject in need of such treatment a fugetactic agent in an amount effective to inhibit endothelial cell migration to the tumor site in the subject.
  • a fugetactic agent in an amount effective to inhibit endothelial cell migration to the tumor site in the subject.
  • the area surrounding the tumor site may not be immediate to the tumor site.
  • Important fugetactic agents are as described above.
  • the method involves administering to a subject in need of such treatment, an anti- fugetactic agent in an amount effective to inhibit germ cell migration in the subject.
  • an anti- fugetactic agent in an amount effective to inhibit germ cell migration in the subject.
  • the anti-fugetactic agent may be as described above.
  • the method involves administering to a subject in need of such treatment a fugetactic agent in an amount effective to inhibit immune cells from migrating close to a germ cell in the subject.
  • the administration may be local to a germ cell-containing site of the subject.
  • fragments of such polypeptide useful according to the invention are only those with fugetactic activity.
  • a person of ordinary skill in the art could easily determine which fragments have such activity by first creating deletions of the full length polypeptides using methods well known in the art, and testing such fragments for their fugetactic activity according to the teachings of the present invention (see, e.g., Examples).
  • SEQ ID NO: 1 is the amino acid sequence of inter- ⁇ -trypsin inhibitor heavy-chain II precursor (ITI H2) (GenBank Acc. No. P 19823).
  • uginosi agents agents with eukaryotic migratory-cell repellant activity
  • the invention involves the unexpected discovery of agents, previously known to be chemoattractants at defined concentrations, to be acting as fugetactic agents at concentrations higher than the defined concentrations.
  • Pharmaceutical compositions containing the foregoing fugetactic agents, and various therapeutic and diagnostic methods utilizing the foregoing fugetactic agents, are also described in more detail below. The foregoing can be used, inter alia, in the treatment of conditions characterized by a need to modulate migratory-cell movement in specific sites in a subject.
  • a subject is a human, non-human primate, cow, horse, pig, sheep, goat, dog, cat or rodent. In all embodiments, human subjects are preferred.
  • fugetactic activity it is meant the ability of an agent to repel (or chemorepel) a eukaryotic cell with migratory capacity (i.e., a cell that can move away from a repellant stimulus). Accordingly, an agent with fugetactic activity is a “fugetactic agent.” Such activity can be detected using any of the transmigration systems described herein (see Examples), or a variety of other systems well known in the art (see, e.g., U.S. patent 5,514,555, entitled: “Assays and therapeutic methods based on lymphocyte chemoattractants,” issued May 7, 1996, to Springer, TA, et al. ).
  • the thymic stromal-cell isolate is useful generally in conditions where inhibition of migration of immune cells to a specific site is desirable. Such conditions are described in greater detail below and in their majority are associated with an inflammatory response.
  • the thymic stromal-cell isolate is produced by thymic stromal cells.
  • the thymic stromal-cell isolate is protease and heat sensitive, and it elutes-off a typical sepharose ion exchange column with 0.5M NaCl at pH 6.3, and wherein the agent is not SDF-1 ⁇ .
  • the thymic stromal-cell isolate may comprise a polypeptide, and it is this polypeptide that repels immune cells.
  • the thymic stromal-cell isolate does not contain an immune cell repelling concentration of SDF-1 ⁇ .
  • An "immune cell repelling concentration of SDF-1 ⁇ ,” as used herein, is any concentration of SDF-1 ⁇ higher than about 100ng/ml.
  • An immune cell repelling concentration of SDF-la may also be any concentration of SDF-1 ⁇ higher than about 150ng/ml.
  • An immune cell repelling concentration of SDF-1 ⁇ may also be any concentration of SDF-1 ⁇ higher than a concentration selected from the group consisting of about 300ng/ml, 500ng/ml, 750ng/ml, and 1 ⁇ g/ml.
  • isolated means separated from its native environment and present in sufficient quantity to permit its identification or use. Isolated, when referring to a protein or polypeptide, means, for example: (i) selectively produced by expression cloning or (ii) partially purified as by chromatography or electrophoresis. Isolated proteins or polypeptides may, but need not be, substantially pure. The thymic stromal-cell isolate may be substantially pure polypeptide.
  • substantially pure means that the protein(s) or polypeptide(s) is essentially free of other substances with which it (they) may be found in nature or in vitro systems, to an extent practical and appropriate for their intended use.
  • substantially pure polypeptides may be produced by techniques well known in the art. Because an isolated protein may be admixed with a pharmaceutically acceptable carrier in a pharmaceutical preparation, the protein may comprise only a small percentage by weight of the preparation. The protein is nonetheless isolated in that it has been separated from many of the substances with which it may be associated in living systems, i.e. isolated from certain other proteins.
  • the fugetactic polypeptide (or thymic stromal-cell isolate) can be isolated from a non-homogenous proteinaceous solution such as a cell culture supernatant or cell homogenate.
  • Thymic stromal-cells can be isolated from a subject by the disaggregation of a piece of thymic tissue, and forming cell suspensions. These thymic stromal-cell suspensions can be cultured according to standard cell culture techniques. In small scale, the cultures can be contained in culture plates, flasks, and dishes. In larger scale, the cultures can be contained in roller bottles, spinner flasks and other large scale culture vessels such as fermenters. Culturing in a three-dimensional, porous, solid matrix may also be used. In preferred embodiments, thymic stromal-cells are obtained from human fetal thymi of 16 to 22 week abortuses as described in greater detail below.
  • the thymic stromal-cell isolate can be isolated from the supernatants of the above-described cell cultures, although the entire culture can be homogenized and subjected to the steps described below for isolation of a fugetactic polypeptide (thymic stromal-cell isolate).
  • the supernatant is removed by aspiration or by centrifugation of the cell culture to remove the cells.
  • the cultures can also be filtered to remove cells and cell debris.
  • the collected supernatant is (in its entirety) the thymic stromal-cell isolate.
  • the fugetactic polypeptide (thymic stromal-cell isolate)-containing supernatant can be fractionated according to standard chromatographic procedures to facilitate isolation of the fugetactic polypeptide (or thymic stromal-cell isolate).
  • standard chromatographic procedures include, but are not limited to, size-exclusion chromatography, FPLC, HPLC, gel filtration chromatography, ion-exchange chromatography, hydrophobic chromatography, immune-affinity chromatography, etc.
  • the fractions of thymic stromal-cell isolate containing supernatant may be used to repel immune cells, particularly mature T cells.
  • Immune cells as used herein are cells of hematopoietic origin (see later discussion), that are involved in the specific recognition of antigens. Immune cells include antigen presenting cells (APCs), such as dendritic cells or macrophages, B cells, T cells, etc.
  • APCs antigen presenting cells
  • Mature T cells as used herein include T cells of a CD4 lo CD8 hi CD69 + TCR + , CD4 hi CD8 lo CD69 + TCR + , CD4 + CD3 + RO + and/or CD8 + CD3 + RO + phenotype.
  • the fugetactic response of the mature T cells to the thymic stromal-cell isolate can be measured as described above, or according to the transmigration assays described in greater detail in the Examples. Other suitable methods will be known to one of ordinary skill in the art and can be employed using only routine experimentation.
  • the fractions which are positive for the thymic stromal-cell isolate can be subjected to additional rounds of screening using the foregoing methodology.
  • the purity of the fraction can be assessed after each round of culture stimulation by subjecting an aliquot of the fraction to SDS-PAGE or other analytical method for visualizing the mixture of constituents in the fraction.
  • the nature of the thymic stromal-cell isolate as a protein, nucleic acid, lipid, carbohydrate etc., can be confirmed at any time by treating an aliquot of a positive fraction with non-specific degradative enzymes for the foregoing classes of molecules and testing the treated fraction in the same assays detailed above.
  • the thymic stromal-cell isolate can then be further isolated if desired using immunological and molecular biological methods (see, e.g. Molecular Cloning: A Laboratory Manual, J. Sambrook, et al., eds., Second Edition, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 1989 , or Current Protocols in Molecular Biology, F.M. Ausubel, et al., eds., John Wiley & Sons, Inc., New York ).
  • a fraction positive for the thymic stromal-cell isolate i.e. with fugetactic activity
  • fugetactic activity which is sufficiently purified can be subjected to protein sequencing according to standard methods.
  • the fraction can be subjected to SDS-PAGE, transferred to a membrane such as polyvinylidene fluoride by electroblotting, and N-terminal amino sequence determined by Edman degradation.
  • a membrane such as polyvinylidene fluoride by electroblotting
  • Any sequence information can be used to screen databases for homology to existing proteins and also to generate degenerate nucleic acids useful for screening a cDNA library by standard methods such as colony hybridization or polymerase chain reaction.
  • the positive fraction can be used to generate antibodies which recognize the thymic stromal-cell isolate. Such antibodies can then be used in expression cloning protocols, Western blots, and other techniques useful in isolation of the thymic stromal-cell isolate.
  • any cDNA libraries, expression libraries, etc. are preferably created from thymic stromal-cells.
  • the description also makes it possible to isolate proteins which bind to the thymic stromal-cell isolate as disclosed herein, including antibodies and cellular binding partners of the thymic stromal-cell isolate such as receptors.
  • the thymic stromal-cell isolate is isolated according to standard methods known to one of ordinary skill in the art, the thymic stromal-cell isolate (or even a substantially purified cell supernatant or fraction) can be used to generate polyclonal or monoclonal antibodies according to standard methods ( see e.g., Harlow and Lane, eds., Antibodies: A Laboratory Manual, Cold Spring Harbor Press, Cold Spring Harbor, NY, 1988 )
  • the proteins which bind to the thymic stromal-cell isolate can be used, for example, in screening assays to detect the presence or absence of thymic stromal-cell isolate and complexes of thymic stromal-cell isolate and their respective binding partners and in purification protocols to isolate thymic stromal-cell isolate and complexes of thymic stromal-cell isolate and their respective binding partners.
  • the binding proteins also can be used to block the effects of the thymic stromal-cell isolate. Such assays can be used to confirm the specificity of binding.
  • peptide binding agents which, for example, can be antibodies or fragments of antibodies having the ability to selectively bind to thymic stromal-cell isolate and inhibit the fugetactic properties of the isolate or agent (or agent as described above).
  • the peptide binding agents may bind selectively to a substantially pure organic agent that repels mature T-cells.
  • the peptide binding agents may bind selectively to SDF-1 ⁇ and inhibit repulsion of immune cells.
  • Antibodies include polyclonal and monoclonal antibodies, prepared according to conventional methodology.
  • an antibody from which the pFc' region has been enzymatically cleaved, or which has been produced without the pFc' region designated an F(ab') 2 fragment
  • an antibody from which the Fc region has been enzymatically cleaved, or which has been produced without the Fc region designated an Fab fragment
  • Fab fragments consist of a covalently bound antibody light chain and a portion of the antibody heavy chain denoted Fd.
  • the Fd fragments are the major determinant of antibody specificity (a single Fd fragment may be associated with up to ten different light chains without altering antibody specificity) and Fd fragments retain epitope-binding ability in isolation.
  • CDRs complementarity determining regions
  • FRs framework regions
  • CDR1 through CDR3 complementarity determining regions
  • the present invention also provides for F(ab') 2 , Fab, Fv and Fd fragments; chimeric antibodies in which the Fc and/or FR and/or CDR1 and/or CDR2 and/or light chain CDR3 regions have been replaced by homologous human or non-human sequences; chimeric F(ab') 2 fragment antibodies in which the FR and/or CDR1 and/or CDR2 and/or light chain CDR3 regions have been replaced by homologous human or non-human sequences; chimeric Fab fragment antibodies in which the FR and/or CDR1 and/or CDR2 and/or light chain CDR3 regions have been replaced by homologous human or non-human sequences; and chimeric Fd fragment antibodies in which the FR and/or CDR1 and/or CDR2 regions have been replaced by homologous human or non-human sequences.
  • the present invention also includes so-called single chain antibodies.
  • polypeptides of numerous size and type that bind specifically to thymic stromal-cell isolate, and complexes of both thymic stromal-cell isolate and its binding partners.
  • polypeptide binding agents can be provided by degenerate peptide libraries which can be readily prepared in solution, in immobilized form or as phage display libraries.
  • Combinatorial libraries also can be synthesized of peptides containing one or more amino acids. Libraries further can be synthesized of peptoids and non-peptide synthetic moieties.
  • antibodies and other binding molecules may be used for example to identify tissues expressing protein or to purify protein.
  • Antibodies also may be coupled to specific diagnostic labeling agents for imaging of cells and tissues that express a thymic stromal-cell isolate.
  • the invention relates to the unexpected discovery that agents previously known to be acting as chemoattractants of immune cells at defined concentrations, also act as fugetactic agents to the same cells at concentrations higher than the defined concentrations.
  • Such fugetactic agents include cytokines.
  • Cytokine is a generic term for nonantibody soluble proteins which are released from one cell subpopulation and which act as intercellular mediators, for example, in the generation or regulation of an immune response. See Human Cytokines: Handbook for Basic & Clinical Research (Aggrawal, et al. eds., Blackwell Scientific, Boston, Mass. 1991 ). Cytokines include, e.g., interleukins IL-1 through IL-15, tumor necrosis factors ⁇ & ⁇ , interferons ⁇ , ⁇ , and ⁇ , tumor growth factor beta (TGF- ⁇ ), colony stimulating factor (CSF) and granulocyte monocyte colony stimulating factor (GM-CSF).
  • TGF- ⁇ tumor growth factor beta
  • CSF colony stimulating factor
  • GM-CSF granulocyte monocyte colony stimulating factor
  • Cytokines share many properties of hormones, but are distinct from classical hormones in that in vivo, they generally act locally on neighboring cells within a tissue.
  • the activities of cytokines range from promoting cell growth (e.g., IL-2, IL-4, and IL-7), and arresting growth (IL-10, tumor necrosis factor and TGF- ⁇ ), to inducing viral resistance (IFN ⁇ , ⁇ , and ⁇ ).
  • IL-2, IL-4, and IL-7 e.g., IL-4, and IL-7
  • IL-10 e.g., tumor necrosis factor and TGF- ⁇
  • IFN ⁇ , ⁇ , and ⁇ inducing viral resistance
  • the cytokine is a cytokine with chemoattractant and/or chemokinetic properties.
  • cytokines include: PAF, N-formylated peptides, C5a, LTB 4 , LXA 4 , chemokines: CXC, IL-8, GCP-2, GRO ⁇ , GRO ⁇ , GRO ⁇ , ENA-78, NAP-2, IP-10, MIG, I-TAC, SDF-1 ⁇ , BCA-1, PF4, Bolekine, MIP-1 ⁇ , MIP-1 ⁇ , RANTES, HCC-1, MCP-1, MCP-2, MCP-3, MCP-4, MCP-5 (mouse only), Leukotactin-1 (HCC-2, MIP-5), Eotaxin, Eotaxin-2 (MPIF2), Eotaxin-3 (TSC), MDC, TARC, SLC (Exodus-2, 6CKine), MIP-3 ⁇ (LARC, Exodus-1), ELC (MIP
  • Cytokines may be members of the Cys-X-Cys family of chemokines (chemokines that bind to the CXCR-4 receptor). Such agents include SDF-1 ⁇ , SDF-1 ⁇ , and met- SDF-1 ⁇ .
  • the cytokine used in the present invention is SDF-1 ⁇ .
  • Fugetactic agents include other CXCR-4 receptor ligands
  • CXCR-4 ligands include, but are not limited to, HIV-I IIIB gp120, small molecules T134 and MD3100, and/or T22 ([Tyr5,12,Lys7]-polyphemusin II) ( Heveker et al., Curr Biol, 1998, 8:369-76 ).
  • the CXCR-4 ligand used in the present invention is HIV-I IIIB gp120.
  • SDF-1 ⁇ is a cytokine (chemokine) produced by thymic and bone marrow stroma ( 12-15 and U.S. Patent 5,756,084, entitled: "Human stromal derived factor 1 ⁇ and 1 ⁇ ,” issued May 26, 1998, to Honjo, et al. ), that has been reported as a highly efficacious and highly potent lymphocyte chemoattractant at concentrations lower than about 100ng/ml.
  • SDF-1 ⁇ at a concentration higher than about 200ng/ml, and preferably higher than about 1 ⁇ g/ml acts a potent fugetactic agent to immune cells, and more specifically to mature T cells, as described in greater detail in the Examples.
  • HIV-1 IIIB gp120 Recombinant envelope glycoprotein gp120 of substrain IIIB of HIVLAI (HIV-1 IIIB gp120) has been used extensively in the art to generate neutralizing antibodies to HIV. The association between gp120 and the CXCR-4 co-receptor for efficient HIV entry into a cell has also been established ( Stott EJ, et al., J Gen Virol, 1998, 79(Pt3):423-32 ). We have discovered, that HIV-1 IIIB gp120 at a concentration higher than about 10ng/ml, and preferably higher than about 200ng/ml, acts a potent fugetactic agent to immune cells, and more specifically to mature T cells, as described in greater detail in the Examples.
  • a culture of stromal-cells is first prepared.
  • Stromal cells comprising fibroblasts with or without other cells and elements described below (the term "fibroblasts" is used collectively herein to include such cells), are inoculated onto a tissue culture vessel (a typical two-dimensional culture dish or three-dimensional matrix).
  • tissue culture vessel a typical two-dimensional culture dish or three-dimensional matrix.
  • fibroblasts may be derived from tissues or organs that include, but are not limited to, skin, liver, pancreas, bone marrow, lymph node, thymus, kidney, CNS, brain, etc., which can be obtained by biopsy (where appropriate) or upon autopsy. In fact fibroblasts can be obtained in quantity rather conveniently from any appropriate cadaver organ. Fetal fibroblasts can also be used.
  • Fibroblasts may be readily isolated by disaggregating an appropriate organ or tissue which is to serve as the source of the fibroblasts. This may be readily accomplished using techniques known to those skilled in the art.
  • the tissue or organ can be disaggregated mechanically and/or treated with digestive enzymes and/or chelating agents that weaken the connections between neighboring cells, making it possible to disperse the tissue into a suspension of individual cells without appreciable cell breakage.
  • Enzymatic dissociation can be accomplished by mincing the tissue and treating the minced tissue with any of a number of digestive enzymes either alone or in combination.
  • the suspension can be fractionated into subpopulations from which the fibroblasts and/or other stromal cells and/or elements can be obtained.
  • This also may be accomplished using standard techniques for cell separation including, but not limited to, cloning and selection of specific cell types, selective destruction of unwanted cells (negative selection), separation based upon differential cell agglutinability in the mixed population, freeze-thaw procedures, differential adherence properties of the cells in the mixed population, filtration, conventional and zonal centrifugation, centrifugal elutriation (counter-streaming centrifugation), unit gravity separation, countercurrent distribution, electrophoresis and fluorescence-activated cell sorting.
  • the isolation of fibroblasts may, for example, be carried out as follows: fresh tissue samples are thoroughly washed and minced in Hank's balanced salt solution (HBSS) in order to remove serum. The minced tissue is incubated from 1-12 hours in a freshly prepared solution of a dissociating enzyme such as trypsin. After such incubation, the dissociated cells are suspended, pelleted by centrifugation and plated onto culture dishes. All fibroblasts will attach before other cells, therefore, appropriate stromal cells can be selectively isolated and grown. The isolated fibroblasts can then be grown to confluency, lifted from the confluent culture and inoculated onto the three-dimensional matrix (see, Naughton et al., 1987, J.
  • HBSS Hank's balanced salt solution
  • stromal-cells found in loose connective tissue may be inoculated onto the culture vessel along with the fibroblasts.
  • These stromal-cells may readily be derived from the same appropriate organs the fibroblasts are derived, that include, but are not limited to, skin, liver, pancreas, bone marrow, lymph node, thymus, kidney, CNS, brain, etc., using methods known in the art such as those discussed above.
  • Such cells include, but are not limited to, endothelial cells, pericytes, macrophages, monocytes, plasma cells, mast cells, adipocytes, etc.
  • stromal cells of hematopoietic tissue including but not limited to, fibroblasts, endothelial cells, macrophages/monocytes, adipocytes and reticular cells, could be used to inoculate a tissue culture dish and/or a three-dimensional matrix.
  • Hematopoietic stromal-cells may be readily obtained from the "buffy coat" formed in bone marrow suspensions by centrifugation at low forces, e.g., 3000g.
  • Stromal-cells of liver may include fibroblasts, Kupffer cells, and vascular and bile duct endothelial cells.
  • glial cells could be used as the stroma to support the proliferation of neurological cells and tissues; glial cells for this purpose can be obtained by trypsinization or collagenase digestion of embryonic or adult brain ( Ponten and Westermark, 1980, in Federof, S. Hertz, L., eds, Advances in Cellular Neurobiology, Vol. 1, New York, Academic Press, pp.209-227 ).
  • the vessel is incubated in an appropriate nutrient medium under conditions that are metabolically favorable for the growth of the cells.
  • the phrase 'metabolically favorable conditions refers to conditions that promote cell division. Such conditions include growth in nutrient medium at 37°C in a 5% CO 2 incubator with greater than 90% humidity.
  • Many commercially available media such as RPMI 1640, Fisher's, Iscove's, McCoy's, Dulbecco's Modified Eagle's Medium, etc., and the like, which may or may not be supplemented with serum, may be suitable for use as nutrient medium.
  • Antibiotics such as penicillin and streptomycin may also be included.
  • the stromal cells are cultured first, for a brief period of time (e.g., at least 1 hour), in the presence of 10% serum.
  • the medium is then changed to a serum-free medium in order to minimize the number of extraeneous (i.e. nonstromal-cell derived) agents that would be present in the medium under continuous culture.
  • the serum-free stromal-cell medium is therefore going to become a stromal-cell conditioned medium (supernatant) upon the continuous incubationof the stromal-cells.
  • Newly inoculated cultures may be allowed to grow under metabolically favorable conditions for between about 3-6 cell cycles.
  • a cell cycle as defined herein, is the length of time between mitoses.
  • the stromal-cells are grown to confluency (for example, at a concentration of of 10 7 cells/10 ml medium in suspension).
  • the supernatant from the culture of stromal-cells is then isolated. Typically, the supernatant is removed by aspiration or by centrifugation of the cell culture to remove the cells. The cultures can also be filtered to remove cells and cell-debris. The supernatant (which is suspected of containing a fugetactic agent) is then fractionated into a plurality of fractions, and a fraction from the plurality of fractions is contacted with a cell with migratory capacity.
  • a "cell with migratory capacity” is a eukaryotic cell that includes, but is not limited to, a cell of hematopoietic origin, a cell of neural origin, a cell of epithelial origin, a cell involved in angiogenesis, a cell of mesenchymal origin, an embryonic stem cell, or a germ cell.
  • Such cells with migratory capacity can respond to a variety of chemotactic signals, including chemoattractive and chemorepulsive (fugetactic) signals. The response typically involves changes in the actin cytoskeleton.
  • Cells of "hematopoietic origin” include, but are not limited to, pluripotent stem cells, multipotent progenitor cells and/or progenitor cells committed to specific hematopoietic lineages.
  • the progenitor cells committed to specific hematopoietic lineages may be of T cell lineage, B cell lineage, dendritic cell lineage, Langerhans cell lineage and/or lymphoid tissue-specific macrophage cell lineage.
  • the hematopoietic cells may be derived from a tissue such as bone marrow, peripheral blood (including mobilized peripheral blood), umbilical cord blood, placental blood, fetal liver, embryonic cells (including embryonic stem cells), aortal-gonadal-mesonephros derived cells, and lymphoid soft tissue. Lymphoid soft tissue includes the thymus, spleen, liver, lymph node, skin, tonsil and Peyer's patches.
  • the "hematopoietic origin" cells may be derived from in vitro cultures of any of the foregoing cells, and in particular in vitro cultures of progenitor cells.
  • Cells of neural origin include neurons and glia, and/or cells of both central and peripheral nervous tissue that express RR/B (see, U.S. Patent 5,863,744, entitled: “Neural cell protein marker RR/B and DNA encoding same,” issued January 26, 1999, to Avraham, et al. ).
  • Cells of epithelial origin include cells of a tissue that covers and lines the free surfaces of the body.
  • Such epithelial tissue includes cells of the skin and sensory organs, as well as the specialized cells lining the blood vessels, gastrointestinal tract, air passages, ducts of the kidneys and endocrine organs.
  • Cells of mesenchymal origin include cells that express typical fibroblast markers such as collagen, vimentin and fibronectin.
  • Cells involved in angiogenesis are cells that are involved in blood vessel formation and include cells of epithelial origin and cells of mesenchymal origin.
  • An embryonic stem cell is a cell that can give rise to cells of all lineages; it also has the capacity to self-renew.
  • a germ cell is a cell specialised to produce haploid gametes. It is a cell further differentiated than a stem cell, that can still give rise to more differentiated germ-line cells.
  • Movement of the cell with migratory capacity is then measured according to any of the methods decribed elsewhere herein (e.g., see Examples), and a person of ordinary skill in the art may determine whether the movement of the cell with migratory capacity is movement away from the fraction. Such movement of the cell with migratory capacity away from the fraction is indicative of the presence of a fugetactic agent in the fraction.
  • the fugetactic agent (stromal or other -cell isolate)-containing supernatant can be fractionated according to standard chromatographic procedures to facilitate isolation of the fugetactic polypeptide (thymic stromal or other -cell isolate).
  • standard chromatographic procedures include, but are not limited to, size-exclusion chromatography, FPLC, HPLC, gel filtration chromatography, ion-exchange chromatography, hydrophobic chromatography, immune-affinity chromatography, etc. (see also the Examples section).
  • the stromal-cells may be thymic stromal-cells and the cell with migratory capacity is a hematopoietic cell.
  • the stromal-cells may be isolated from an immune-privileged site/tissue.
  • An "immune-privileged tissue” is a tissue with immune-cell tolerance and includes, but is not limited to, brain tissue, central nervous system (CNS), testes, placenta, or ocular tissue (e.g., aqueous humour).
  • the biological fluid includes, but is not limited to, synovial fluid, cerebral spinal fluid, fallopian tube fluid, seminal fluid, ocular fluid, pericardial fluid, pleural fluid, inflammatory exudate and ascitic fluid.
  • the agent suspected of being a fugetactic agent may be an agent present in a tumor cell culture supernatant, tumor cell eluate, and/or tumor cell lysate (see also the Examples section).
  • the tumor cell may be of a cancer or tumor type thought to escape immune recognition.
  • Such cancers or tumors may be of the folowing origin: biliary tract cancer; brain cancer, including glioblastomas and medulloblastomas; breast cancer; cervical cancer; choriocarcinoma; colon cancer; endometrial cancer; esophageal cancer; gastric cancer; hematological neoplasms, including acute lymphocytic and myelogenous leukemia; multiple myeloma; AIDS associated leukemias and adult T-cell leukemia lymphoma; intraepithelial neoplasms, including Bowen's disease and Paget's disease; liver cancer (hepatocarcinoma); lung cancer; lymphomas, including Hodgkin's disease and lymphocytic lymphomas; neuroblastomas; oral cancer, including squamous cell carcinoma; ovarian cancer, including those arising from epithelial cells, stromal cells, germ cells and mesenchymal cells; pancreas cancer; prostate cancer; rectal cancer
  • the cell with migratory capacity is a hematopoietic cell.
  • the hematopoietic cell may be an immune cell.
  • Electrode surfaces include, but are not limited to, dental and orthopedic prosthetic implants, artificial valves, and organic implantable tissue such as a stent, allogeneic and/or xenogeneic tissue, organ and/or vasculature.
  • Implantable prosthetic devices have been used in the surgical repair or replacement of internal tissue for many years.
  • Orthopedic implants include a wide variety of devices, each suited to fulfill particular medical needs. Examples of such devices are hip joint replacement devices, knee joint replacement devices, shoulder joint replacement devices, and pins, braces and plates used to set fractured bones.
  • Some contemporary orthopedic and dental implants use high performance metals such as cobalt-chrome and titanium alloy to achieve high strength. These materials are readily fabricated into the complex shapes typical of these devices using mature metal working techniques including casting and machining.
  • the material surface is coated with an amount of a fugetactic agent effective to repel immune cells.
  • the material surface is part of an implant.
  • the material surface in addition to a fugetactic agent, may also be coated with a cell-growth potentiating agent, an anti-infective agent, and/or an antiinflammatory agent.
  • a cell-growth potentiating agent as used herein is an agent which stimulates growth of a cell and includes growth factors such as PDGF, EGF, FGF, TGF, NGF, CNTF, and GDNF.
  • An anti-infectious agent as used herein is an agent which reduces the activity of or kills a microorganism and includes: Aztreonam; Chlorhexidine Gluconate; Imidurea; Lycetamine; Nibroxane; Pirazmonam Sodium; Propionic Acid; Pyrithione Sodium; Sanguinarium Chloride; Tigemonam Dicholine; Acedapsone; Acetosulfone Sodium; Alamecin; Alexidine; Amdinocillin; Amdinocillin Pivoxil; Amicycline; Amifloxacin; Amifloxacin Mesylate; Amikacin; Amikacin Sulfate; Aminosalicylic acid; Aminosalicylate sodium; Amoxicillin; Amphomycin; Ampicillin; Ampicillin Sodium; Apalcillin Sodium; Apramycin; Aspartocin; Astromicin Sulfate; Avilamycin; Avoparcin; Azithromycin; Azlocillin
  • Antiinflammatory agents are as described below.
  • a method of inhibiting migration of immune cells to a specific site in a subject involves locally administering to a specific site in a subject in need of such treatment a fugetactic agent in an amount effective to inhibit migration of immune cells to the specific site in a subject.
  • Inflammation is a localised protective response elicited by a foreign (non-self) antigen, and/or by an injury or destruction of tissue(s), which serves to destroy, dilute or sequester the foreign antigen, the injurious agent, and/or the injured tissue. Inflammation occurs when tissues are injured by viruses, bacteria, trauma, chemicals, heat, cold, or any other harmful stimuli.
  • T cells classic weapons of the immune system
  • B cells macrophages
  • soluble products that are mediators of inflammatory responses (neutrophils, eosinophils, basophils, kinin and coagulation systems, and complement cascade).
  • a typical inflammatory response is characterized by (i) migration of leukocytes at the site of antigen (injury) localization; (ii) specific and nonspecific recognition of "foreign” and other (necrotic/injured tissue) antigens mediated by B and T lymphocytes, macrophages and the alternative complement pathway; (iii) amplification of the inflammatory response with the recruitment of specific and nonspecific effector cells by complement components, lymphokines and monokines, kinins, arachidonic acid metabolites, and mast cell/basophil products; and (iv) macrophage, neutrophil and lymphocyte participation in antigen destruction with ultimate removal of antigen particles (injured tissue) by phagocytosis.
  • the ability of the immune system to discriminate between "self' and "non-self'(foreign) antigens is therefore vital to the functioning of the immune system as a specific defense against "non-self” antigens.
  • Non-self antigens are those antigens on substances entering a subject, or exist in a subject but are detectably different or foreign from the subject's own constituents
  • self antigens are those which, in the healthy subject, are not detectably different or foreign from its own constituents.
  • an individual's immune system will identify its own constituents as “non-self,” and initiate an immune response against "self-antigens,” at times causing more damage or discomfort as from, for example, an invading microbe or foreign material, and often producing serious illness in a subject.
  • the inflammation may be caused by an immune response against "self-antigen," and the subject in need of treatment according to the invention has an autoimmune disease.
  • "Autoimmune disease” as used herein results when a subject's immune system attacks its own organs or tissues, producing a clinical condition associated with the destruction of that tissue, as exemplified by diseases such as rheumatoid arthritis, uveitis, insulin-dependent diabetes mellitus, hemolytic anemias, rheumatic fever, Crohn's disease, Guillain-Barre syndrome, psoriasis, thyroiditis, Graves' disease, myasthenia gravis, glomerulonephritis, autoimmune hepatitis, multiple sclerosis, systemic lupus erythematosus, etc.
  • Autoimmune disease may be caused by a genetic predisposition alone, by certain exogenous agents (e.g., viruses, bacteria, chemical agents, etc.), or both.
  • Some forms of autoimmunity arise as the result of trauma to an area usually not exposed to lymphocytes, such as neural tissue or the lens of the eye. When the tissues in these areas become exposed to lymphocytes, their surface proteins can act as antigens and trigger the production of antibodies and cellular immune responses which then begin to destroy those tissues.
  • Other autoimmune diseases develop after exposure of a subject to antigens which are antigenically similar to, that is cross-reactive with, the subject's own tissue.
  • rheumatic fever for example, an antigen of the streptococcal bacterium, which causes rheumatic fever, is cross-reactive with parts of the human heart.
  • the antibodies cannot differentiate between the bacterial antigens and the heart muscle antigens, consequently cells with either of those antigens can be destroyed.
  • autoimmune diseases for example, insulin-dependent diabetes mellitus (involving the destruction of the insulin producing beta-cells of the islets of Langerhans), multiple sclerosis (involving the destruction of the conducting fibers of the nervous system) and rheumatoid arthritis (involving the destruction of the joint-lining tissue), are characterized as being the result of a mostly cell-mediated autoimmune response and appear to be due primarily to the action of T cells (See, Sinha et al., Science, 1990, 248:1380 ). Yet others, such as myesthenia gravis and systemic lupus erythematosus, are characterized as being the result of primarily a humoral autoimmune response.
  • inhibition of migration of immune cells to a specific site of inflammation involved in any of the foregoing conditions according to the invention is beneficial to the subject since it inhibits escalation of the inflammatory response, protecting the specific site (e.g., tissue) involved, from "self-damage.”
  • the subject has rheumatoid arthritis, multiple sclerosis, or uveitis.
  • the inflammation may be caused by an immune response against "non-self-antigens" (including antigens of necrotic self-material), and the subject in need of treatment according to the invention is a transplant recipient, has atherosclerosis, has suffered a myocardial infarction and/or an ischemic stroke, has an abscess, and/or has myocarditis.
  • non-self-antigens including antigens of necrotic self-material
  • the method involves locally administering to a specific site in a subject in need of such treatment an anti-fugetactic agent in an amount effective to inhibit immune cell-specific fugetactic activity at a specific site in the subject.
  • the specific site may be a site of a pathogenic infection. Efficient recruitment of immune cells to help eliminate the infection is therefore beneficial.
  • the specific site may be a germ cell containing site. In this case the recruitment of immune cells to these specific sites will help eliminate unwanted germ cells, and/or implanted and nonimplanted embryos. Co-administration of contraceptive agents other than anti-fugetactic agents is also described herein. Non-anti-fugetactic contraceptive agents are well known in the art.
  • the specific site is an area immediately surrounding a tumor. Since most of the known tumors escape immune recognition, it is beneficial to enhance the migration of immune cells to the tumor site.
  • co-administration of anti-cancer agents other than anti-fugetactic agents is also provided.
  • Non-anti-fugetactic anti-cancer agents include: Acivicin; Aclarubicin; Acodazole Hydrochloride; Acronine; Adozelesin; Aldesleukin; Altretamine; Ambomycin; Ametantrone Acetate; Aminoglutethimide; Amsacrine; Anastrozole; Anthramycin; Asparaginase; Asperlin; Azacitidine; Azetepa; Azotomycin; Batimastat; Benzodepa; Bicalutamide; Bisantrene Hydrochloride; Bisnafide Dimesylate; Bizelesin; Bleomycin Sulfate; Brequinar Sodium; Bropirimine; Busulfan; Cactinomycin; Calusterone; Caracemide; Carbetimer; Carboplatin; Carmustine; Carubicin Hydrochloride; Carzelesin; Cedefingol; Chlorambucil; Cirolemycin; Cis
  • the method involves locally administering to a tumor site in a subject in need of such treatment an anti-fugetactic agent in an amount effective to inhibit metastasis of tumor cells from the tumor site in the subject.
  • Anti-cancer agents are as described above.
  • the method involves locally administering to an area surrounding a tumor site in a subject in need of such treatment a fugetactic agent in an amount effective to inhibit endothelial cell migration to the tumor site in the subject.
  • the area surrounding the tumor site may not be immediate to the tumor site.
  • Important fugetactic agents are as described above.
  • the method involves administering to a subject in need of such treatment, an anti-fugetactic agent in an amount effective to inhibit migration of germ cells in the subject.
  • the administration may be local to a germ cell-containing site of the subject.
  • the method involves administering to a subject in need of such treatment a fugetactic agent in an amount effective to inhibit immune cells from migrating close to a germ cell (including an egg, a sperm, a fertilized egg, or an implanted embryo) in the subject.
  • a fugetactic agent in an amount effective to inhibit immune cells from migrating close to a germ cell (including an egg, a sperm, a fertilized egg, or an implanted embryo) in the subject.
  • the administration may be local to a germ cell-containing site of the subject.
  • the foregoing methods of therapy may include co-administration of a non-fugetactic agent together with a fugetactic agent of the invention that can act cooperatively, additively, or synergistically with the fugetactic agent of the invention to inhibit migration of immune cells to the site of inflammation in the subject.
  • a fugetactic agent may be administered substantially simultaneously with a non-fugetactic agent to inhibit migration of immune cells to a site of inflammation.
  • substantially simultaneously it is meant that the fugetactic agent is locally administered to the subject close enough in time with the administration of the non-fugetactic agent, whereby the non-fugetactic agent may exert a potentiating effect on migration inhibiting activity of the fugetactic agent.
  • the fugetactic agent is administered before, at the same time, and/or after the administration of the non-fugetactic agent.
  • the fugetactic agent can be administered as a polypeptide, and/or a nucleic acid which expresses a fugetactic agent.
  • the non-fugetactic agents may be immunosuppressants.
  • immunosuppressants include: Azathioprine; Azathioprine Sodium; Cyclosporine; Daltroban; Gusperimus Trihydrochloride; Sirolimus; Tacrolimus.
  • the non-fugetactic agents may be anti-inflammatory agents.
  • anti-inflammatory agents include: Alclofenac; Alclometasone Dipropionate; Algestone Acetonide; Alpha Amylase; Amcinafal; Amcinafide; Amfenac Sodium; Amiprilose Hydrochloride; Anakinra; Anirolac; Anitrazafen; Apazone; Balsalazide Disodium; Bendazac; Benoxaprofen; Benzydamine Hydrochloride; Bromelains; Broperamole; Budesonide; Carprofen; Cicloprofen; Cintazone; Cliprofen; Clobetasol Propionate; Clobetasone Butyrate; Clopirac; Cloticasone Propionate; Cormethasone Acetate; Cortodoxone; Deflazacort; Desonide; Desoximetasone; Dexamethasone Dipropionate; Diclofenac Potassium; Diclofenac
  • compositions as described above, are administered in effective amounts.
  • the effective amount will depend upon the mode of administration, the particular condition being treated and the desired outcome. It will also depend upon, as discussed above, the stage of the condition, the age and physical condition of the subject, the nature of concurrent therapy, if any, and like factors well known to the medical practitioner. For therapeutic applications, it is that amount sufficient to achieve a medically desirable result. In some cases this is a local (site-specific) reduction of inflammation. In other cases, it is inhibition of tumor growth and/or metastasis.
  • doses of active compounds of the present invention would be from about 0.01 mg/kg per day to 1000 mg/kg per day. It is expected that doses ranging from 50-500 mg/kg will be suitable.
  • a variety of administration routes are available. The methods of the invention, generally speaking, may be practiced using any mode of administration that is medically acceptable, meaning any mode that produces effective levels of the active compounds without causing clinically unacceptable adverse effects. Such modes of administration include oral, rectal, topical, nasal, interdermal, or parenteral routes.
  • parenteral includes subcutaneous, intravenous, intramuscular, or infusion. Intravenous or intramuscular routes are not particularly suitable for long-term therapy and prophylaxis. They could, however, be preferred in emergency situations.
  • Oral administration will be preferred for prophylactic treatment because of the convenience to the patient as well as the dosing schedule.
  • a desirable route of administration is by pulmonary aerosol.
  • Techniques for preparing aerosol delivery systems containing peptides are well known to those of skill in the art. Generally, such systems should utilize components which will not significantly impair the biological properties of the antibodies, such as the paratope binding capacity (see, for example, Sciarra and Cutie, "Aerosols," in Remington's Pharmaceutical Sciences, 18th edition, 1990, pp 1694-1712 ).
  • those of skill in the art can readily determine the various parameters and conditions for producing antibody or peptide aerosols without resort to undue experimentation.
  • compositions suitable for oral administration may be presented as discrete units, such as capsules, tablets, lozenges, each containing a predetermined amount of the active agent.
  • Other compositions include suspensions in aqueous liquids or non-aqueous liquids such as a syrup, elixir or an emulsion.
  • Preparations for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions, and emulsions.
  • non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate.
  • Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media.
  • Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's or fixed oils.
  • Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers (such as those based on Ringer's dextrose), and the like. Preservatives and other additives may also be present such as, for example, antimicrobials, anti-oxidants, chelating agents, and inert gases and the like. Lower doses will result from other forms of administration, such as intravenous administration. In the event that a response in a subject is insufficient at the initial doses applied, higher doses (or effectively higher doses by a different, more localized delivery route) may be employed to the extent that patient tolerance permits. Multiple doses per day are contemplated to achieve appropriate systemic levels of compounds.
  • the fugetactic agents, fugetactic binding agents, or fragments thereof may be combined, optionally, with a pharmaceutically-acceptable carrier.
  • pharmaceutically-acceptable carrier means one or more compatible solid or liquid filler, diluents or encapsulating substances which are suitable for administration into a human.
  • carrier denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application.
  • the components of the pharmaceutical compositions also are capable of being co-mingled with the molecules of the present invention, and with each other, in a manner such that there is no interaction which would substantially impair the desired pharmaceutical efficacy.
  • compositions of fugetactic agents and anti-fugetactic agents are also described herein pharmaceutical compositions of fugetactic agents and anti-fugetactic agents.
  • the pharmaceutical preparations described herein are applied in pharmaceutically-acceptable amounts and in pharmaceutically-acceptably compositions.
  • Such preparations may routinely contain salt, buffering agents, preservatives, compatible carriers, and optionally other therapeutic agents.
  • the salts should be pharmaceutically acceptable, but non-pharmaceutically acceptable salts may conveniently be used to prepare pharmaceutically-acceptable salts thereof and are not excluded from the scope of the invention.
  • Such pharmacologically and pharmaceutically-acceptable salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulfuric, nitric, phosphoric, maleic, acetic, salicylic, citric, formic, malonic, succinic, and the like.
  • pharmaceutically-acceptable salts can be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium or calcium salts.
  • Fugetactic molecules preferably are produced recombinantly, although such molecules may be isolated from biological extracts.
  • Recombinantly produced fugetactic agents such as SDF-1 ⁇ polypeptides, include chimeric proteins comprising a fusion of a SDF-1 ⁇ protein with another polypeptide, e.g., a polypeptide capable of providing or enhancing protein-protein binding, sequence specific nucleic acid binding (such as GAL4), enhancing stability of the SDF-1 ⁇ polypeptide under assay conditions, or providing a detectable moiety, such as green fluorescent protein.
  • a polypeptide fused to a SDF-1 ⁇ polypeptide or fragment may also provide means of readily detecting the fusion protein, e.g., by immunological recognition or by fluorescent labeling.
  • nucleic acids of the invention SDF-1 ⁇ sense and anti-sense, dominant negative
  • Such techniques include transfection of nucleic acid-CaPO 4 precipitates, transfection of nucleic acids associated with DEAE, transfection with a retrovirus including the nucleic acid of interest, liposome mediated transfection, and the like.
  • a vehicle used for delivering a nucleic acid of the invention into a cell e.g., a retrovirus, or other virus; a liposome
  • a molecule such as an antibody specific for a surface membrane protein on the target cell or a ligand for a receptor on the target cell can be bound to or incorporated within the nucleic acid delivery vehicle.
  • proteins which bind to a surface membrane protein associated with endocytosis may be incorporated into the liposome formulation for targeting and/or to facilitate uptake.
  • proteins include capsid proteins or fragments thereof tropic for a particular cell type, antibodies for proteins which undergo internalization in cycling, proteins that target intracellular localization and enhance intracellular half life, and the like.
  • Polymeric delivery systems also have been used successfully to deliver nucleic acids into cells, as is known by those skilled in the art. Such systems even permit oral delivery of nucleic acids.
  • Other delivery systems can include time-release, delayed release or sustained release delivery systems. Such systems can avoid repeated administrations of the fugetactic agent, increasing convenience to the subject and the physician.
  • Many types of release delivery systems are available and known to those of ordinary skill in the art. They include polymer base systems such as poly(lactide-glycolide), copolyoxalates, polycaprolactones, polyesteramides, polyorthoesters, polyhydroxybutyric acid, and polyanhydrides. Microcapsules of the foregoing polymers containing drugs are described in, for example, U.S. Patent 5,075,109 .
  • Delivery systems also include non-polymer systems that are: lipids including sterols such as cholesterol, cholesterol esters and fatty acids or neutral fats such as mono- di- and tri-glycerides; hydrogel release systems; sylastic systems; peptide based systems; wax coatings; compressed tablets using conventional binders and excipients; partially fused implants; and the like.
  • lipids including sterols such as cholesterol, cholesterol esters and fatty acids or neutral fats such as mono- di- and tri-glycerides
  • hydrogel release systems such as cholesterol, cholesterol esters and fatty acids or neutral fats such as mono- di- and tri-glycerides
  • sylastic systems such as cholesterol, cholesterol esters and fatty acids or neutral fats such as mono- di- and tri-glycerides
  • peptide based systems such as fatty acids
  • wax coatings such as those described in U.S. Patent Nos.
  • a preferred delivery system is a colloidal dispersion system.
  • Colloidal dispersion systems include lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes.
  • a preferred colloidal system is a liposome.
  • Liposomes are artificial membrane vessels which are useful as a delivery vector in vivo or in vitro. It has been shown that large unilamellar vessels (LUV), which range in size from 0.2 - 4.0 ⁇ m can encapsulate large macromolecules. RNA, DNA, and intact virions can be encapsulated within the aqueous interior and be delivered to cells in a biologically active form ( Fraley, et al., Trends Biochem. Sci., (1981) 6:77 ).
  • LUV large unilamellar vessels
  • a liposome In order for a liposome to be an efficient gene transfer vector, one or more of the following characteristics should be present: (1) encapsulation of the gene of interest at high efficiency with retention of biological activity; (2) preferential and substantial binding to a target cell in comparison to non-target cells; (3) delivery of the aqueous contents of the vesicle to the target cell cytoplasm at high efficiency; and (4) accurate and effective expression of genetic information.
  • Liposomes may be targeted to a particular tissue by coupling the liposome to a specific ligand such as a monoclonal antibody, sugar, glycolipid, or protein.
  • a specific ligand such as a monoclonal antibody, sugar, glycolipid, or protein.
  • Liposomes are commercially available from Gibco BRL, for example, as LIPOFECTINTM and LIPOFECTACETM, which are formed of cationic lipids such as N-[1-(2, 3 dioleyloxy)-propyl]-N, N, N-trimethylammonium chloride (DOTMA) and dimethyl dioctadecylammonium bromide (DDAB).
  • DOTMA N-[1-(2, 3 dioleyloxy)-propyl]-N, N, N-trimethylammonium chloride
  • DDAB dimethyl dioctadecylammonium bromide
  • the preferred vehicle may be a biocompatible microparticle or implant that is suitable for implantation into the mammalian recipient.
  • Exemplary bioerodible implants that are useful in accordance with this method are described in PCT International application no. PCT/US/03307 (Publication No. WO 95/24929 , entitled “Polymeric Gene Delivery System”).
  • PCT/US/0307 describes a biocompatible, preferably biodegradable polymeric matrix for containing an exogenous gene under the control of an appropriate promoter. The polymeric matrix is used to achieve sustained release of the exogenous gene in the patient.
  • the fugetactic agents described herein are encapsulated or dispersed within the biocompatible, preferably biodegradable polymeric matrix disclosed in PCT/US/03307 .
  • the polymeric matrix preferably is in the form of a microparticle such as a microsphere (wherein a fugetactic agent is dispersed throughout a solid polymeric matrix) or a microcapsule (wherein a fugetactic agent is stored in the core of a polymeric shell).
  • a microparticle such as a microsphere (wherein a fugetactic agent is dispersed throughout a solid polymeric matrix) or a microcapsule (wherein a fugetactic agent is stored in the core of a polymeric shell).
  • Other forms of the polymeric matrix for containing a fugetactic agent include films, coatings, gels, implants, and stents.
  • the size and composition of the polymeric matrix device is selected to result in favorable release kinetics in the tissue into which the matrix is introduced.
  • the size of the polymeric matrix further is selected according to the method of delivery which is to be used.
  • the polymeric matrix and fugetactic agent are encompassed in a surfactant vehicle.
  • the polymeric matrix composition can be selected to have both favorable degradation rates and also to be formed of a material which is bioadhesive, to further increase the effectiveness of transfer.
  • the matrix composition also can be selected not to degrade, but rather, to release by diffusion over an extended period of time.
  • the delivery system may be a biocompatible microsphere that is suitable for local, site-specific delivery.
  • microspheres are disclosed in Chickering et al., Biotech. And Bioeng., (1996) 52:96-101 and Mathiowitz et al., Nature, (1997) 386:.410-414 .
  • Both non-biodegradable and biodegradable polymeric matrices can be used to deliver the fugetactic agents of the invention to the subject.
  • Biodegradable matrices are preferred.
  • Such polymers may be natural or synthetic polymers. Synthetic polymers are preferred.
  • the polymer is selected based on the period of time over which release is desired, generally in the order of a few hours to a year or longer. Typically, release over a period ranging from between a few hours and three to twelve months is most desirable.
  • the polymer optionally is in the form of a hydrogel that can absorb up to about 90% of its weight in water and further, optionally is cross-linked with multi-valent ions or other polymers.
  • fugetactic agents are delivered using a bioerodible implant by way of diffusion, or more preferably, by degradation of the polymeric matrix.
  • exemplary synthetic polymers which can be used to form the biodegradable delivery system include: polyamides, polycarbonates, polyalkylenes, polyalkylene glycols, polyalkylene oxides, polyalkylene terepthalates, polyvinyl alcohols, polyvinyl ethers, polyvinyl esters, poly-vinyl halides, polyvinylpyrrolidone, polyglycolides, polysiloxanes, polyurethanes and co-polymers thereof, alkyl cellulose, hydroxyalkyl celluloses, cellulose ethers, cellulose esters, nitro celluloses, polymers of acrylic and methacrylic esters, methyl cellulose, ethyl cellulose, hydroxypropyl cellulose, hydroxy-propyl methyl cellulose, hydroxybutyl methyl cellulose, cellulose acetate,
  • non-biodegradable polymers examples include ethylene vinyl acetate, poly(meth)acrylic acid, polyamides, copolymers and mixtures thereof.
  • Bioadhesive polymers of particular interest include bioerodible hydrogels described by H.S. Sawhney, C.P. Pathak and J.A. Hubell in Macromolecules, (1993) 26:581-587 , the teachings of which are incorporated herein, polyhyaluronic acids, casein, gelatin, glutin, polyanhydrides, polyacrylic acid, alginate, chitosan, poly(methyl methacrylates), poly(ethyl methacrylates), poly(butylmethacrylate), poly(isobutyl methacrylate), poly(hexylmethacrylate), poly(isodecyl methacrylate), poly(lauryl methacrylate), poly(phenyl methacrylate), poly(methyl acrylate), poly(isopropyl acrylate), poly(isobutyl acrylate), and poly(octadecyl acrylate).
  • Such implantable pumps include controlled-release microchips.
  • a preferred controlled-release microchip is described in Santini, JT Jr., et al., Nature, 1999, 397:335-338 .
  • Long-term sustained release means that the implant is constructed and arranged to delivery therapeutic levels of the active ingredient for at least 30 days, and preferably 60 days.
  • Long-term sustained release implants are well-known to those of ordinary skill in the art and include some of the release systems described above.
  • the isolated fugetactic agents described herein may be directly to the site at which there is inflammation, e.g., the joints in the case of a subject with rheumatoid arthritis, the blood vessels of an atherosclerotic organ, etc.
  • this can be accomplished by attaching an isolated fugetactic molecule (nucleic acid or polypeptide) to the surface of a balloon catheter; inserting the catheter into the subject until the balloon portion is located at the site of inflammation, e.g. an atherosclerotic vessel, and inflating the balloon to contact the balloon surface with the vessel wall at the site of the occlusion.
  • the compositions can be targeted locally to particular inflammatory sites to modulate immune cell migration to these sites.
  • the local administration involves an implantable pump to the site in need of such treatment.
  • Preferred pumps are as described above.
  • the fugetactic agent may be delivered topically, e.g., in an ointment/dermal formulation.
  • the fugetactic molecules of the invention are delivered in combination with a non-fugetactic molecule (e.g., antiinflammatory, immunosuppressant, etc.).
  • the isolated fugetactic agents are administered to a subject in combination with a balloon angioplasty procedure.
  • a balloon angioplasty procedure involves inserting a catheter having a deflated balloon into an artery.
  • the deflated balloon is positioned in proximity to the atherosclerotic plaque and the site of inflammation, and is inflated such that the plaque is compressed against the arterial wall.
  • the layer of endothelial cells on the surface of the artery is disrupted, thereby exposing the underlying vascular smooth muscle cells.
  • the isolated fugetactic molecule is attached to the balloon angioplasty catheter in a manner which permits release of the isolated fugetactic molecule at the site of the atherosclerotic plaque and the site of inflammation.
  • the isolated fugetactic molecule may be attached to the balloon angioplasty catheter in accordance with standard procedures known in the art.
  • the isolated fugetactic molecule may be stored in a compartment of the balloon angioplasty catheter until the balloon is inflated, at which point it is released into the local environment.
  • the isolated fugetactic molecule may be impregnated on the balloon surface, such that it contacts the cells of the arterial wall as the balloon is inflated.
  • the fugetactic molecule also may be delivered in a perforated balloon catheter such as those disclosed in Flugelman, et al., Circulation, v. 85, p. 1110-1117 (1992 ).
  • Peripheral blood was obtained from 12 healthy donors (age 25- 40 years, median age 31).
  • Peripheral blood mononuclear cells (PBMC) were obtained from peripheral blood using Ficoll-Hypaque.
  • PBMC peripheral blood mononuclear cells
  • PBMC peripheral blood mononuclear cells
  • PBMC peripheral blood mononuclear cells
  • FITC anti-CD45RA
  • FITC anti-CD45RO
  • the CD4 + CD45RA + , CD4 + CD45RO + and CD8 + CD45RA + or CD8 + CD45RO + subpopulations of peripheral blood cells were sorted using a FACS vantage sorter (Becton-Dickinson, San Jose, CA).
  • each T-cell subpopulation was determined to be greater than 99% by immunophenotyping.
  • Chemokine receptor expression including mean fluorescent intensities for chemokine receptors were also measured and compared for each T-cell subpopulation by staining sorted cells with anti-CXCR4 (PE) (Pharmagen/Becton Dickinson, San Diego, CA) directed against the cellular receptor for SDF-1 ⁇ . Sorted adult peripheral blood T-cell subpopulations were cultured overnight in serum- free Iscoves modified medium (GIBCO BRL, Gaithersburg, MD), prior to their use in chemotaxis assays.
  • PE anti-CXCR4
  • Thymi Human fetal thymi and peripheral blood samples were obtained from 16 to 22 week abortuses according to Institutional Review Board approved guidelines. Thymi were physically disaggregated and mononuclear cell suspensions generated. Thymocytes were stained with CD8 (FITC), CD3 (PE) and CD4 (APC) and sorted into purified subpopulations of triple negative immature thymocytes (TN) (CD3 - , CD4 - , CD8 - ) and mature double positive (DP) (CD4 + CD8 + ) and single positive (SP) thymocytes (CD3 + CD4 + or CD3 + CD8 + ) by cell sorting.
  • FITC CD8
  • PE CD3
  • APC CD4
  • FBMC Fetal blood mononuclear cells
  • FITC CD8
  • PE CD3
  • APC CD4
  • FITC anti-CD45RA
  • Purified populations of CD4 + CD45RA + and CD8 + CD45RA + T-cells were sorted cultured overnight in serum free Iscoves modified medium prior to use in chemotaxis assays.
  • chemotaxis assays were used in this study.
  • quantitative chemotaxis transmigration assays were performed using a transwell system (Coming Inc. , NY) (6.5mm diameter, 5 ⁇ m pore size, polycarbonate membrane) as previously described 13 .
  • Purified T-cell and thymocyte subpopulations (5 x 10 4 cells) were added to the upper chamber of each well in a total volume of 150 ⁇ l of IMDM alone.
  • SDF-1 ⁇ (PeproTech, Rocky Hill, NJ) was used at concentrations of 10 ⁇ g/ml, 1 ⁇ g/ml, 100 ng/ml, 10 ng/ml and 0 ng/ml in the lower, upper or both lower and upper chambers of the transwell to generate a checkerboard analysis matrix of positive, negative and absent gradients of SDF-1 ⁇ respectively.
  • PTX pertussis toxin
  • genistein 1 ⁇ g/ml for 20minutes
  • herbimycin 1 ⁇ M for 20 minutes
  • wortmannin 1 ⁇ M for 20 minutes
  • Transwells prepared in this way were incubated for 3 hours at 37°C, 5% CO 2 incubator. Cells were harvested from the lower chamber after 3 hours and cell counts, immunophenotype and CXCR-4 chemokine receptor expression determined.
  • the second semi-quantitative transmigration assay involved the use of a tissue culture insert (6.5 mm diameter, 0.45 ⁇ m pore size, polycarbonate membrane) (Millipore, Bedford MA).
  • a tissue culture insert 6.5 mm diameter, 0.45 ⁇ m pore size, polycarbonate membrane
  • Adult peripheral blood T-cell subpopulations (1 ⁇ 10 5 cells) were plated into the wells of a 12-well tissue-culture dish in serum-free Iscove's medium. The tissue-culture plate was then incubated at 37°C for 1 hour to allow the cells to settle evenly over the base of the well.
  • IMDM containing SDF-1 ⁇ at final concentrations of 10 ⁇ g/ml, 1 ⁇ g/ml, 100 ng/ml, 10 ng/ml or 0ng/ml was then dispensed into the tissue culture insert.
  • the tissue culture insert was gently placed in the center of the well of the 12-well tissue-culture plate leaving a gap of 0.3 cm between the edge of the insert and the edge of the well of the tissue culture plate.
  • the well with transwell insert was incubated for up to 18 hours at 37°C on the heated stage of a microscope and viewed by time lapse videomicroscopy recording at 120 frames per hour or by Polaroid still photography. Maintenance of a gradient was assessed by loading of methylene blue into the insert and observing a preserved gradient of color over the incubation period.
  • T-cell subpopulations (1 x 10 5 cells) were mixed with 0.5 ml of 0.5% methylcellulose (Stem Cell Technologies, Vancouver, BC) in sterile medium and plated in a 24 well plate. 5 ⁇ l of PBS or SDF-1 ⁇ at concentrations of 10 ⁇ g/ml or 100ng/ml was then dispensed gently into the methylcellulose at a set position on the edge of the well prior to incubation for up to 18 hours on a heated (37°C) microscope stage and viewed by time lapse videomicroscopy.
  • Transmigration assays Use of Thymic Stromal-Cell Medium(TSCM)
  • the chemotaxis assays used in this study were as for the SDF-1 ⁇ described above. Quantitative chemotaxis transmigration assays were performed also using a transwell system. Serum free human fetal thymic stroma conditioned medium was added to the top, bottom or top and bottom chambers of the transwell in order to assess the chemotactic activities within the conditioned medium at dilutions of 1:2, 1:10 and 1:15 in serum free IMDM. Heat-inactivated TSCM was also used in the chemotaxis assay.
  • Cell migration was allowed to occur for 3 hours at 37°C in an incubator (5% CO 2 ). Cells were harvested from the lower chamber after 3 hours and accurate cell counts, immunophenotypes, and chemokine receptor expression were determined for the cells which had emigrated from the upper chamber to the lower chamber.
  • Transmigration assays Use of SDF-1, recombinant HIV-1 IIIB gp 120 and inhibitors.
  • chemotaxis assays Two chemotaxis assays were used in this study. First, quantitative chemotaxis transmigration assays were performed as for the SDF-1 ⁇ described above (and Kim, et al., Blood, 1998, 91:4434 ). Purified T-cell and thymocyte subpopulations (5 x 10 4 cells) were added to the upper chamber of each well in a total volume of 150 ⁇ l of IMDM alone.
  • SDF-1 ⁇ (PeproTech, Rocky Hill, NJ) was used at concentrations of 10 ⁇ g/ml, 1 ⁇ g/ml, 100 ng/ml, 10 ng/ml and 0 ng/ml in the lower, upper or both lower and upper chambers of the transwell to generate a checkerboard analysis matrix of positive, negative and absent gradients of SDF-1, respectively.
  • Recombinant endotoxin free HIV-1 IIIB gp 120 (Intracel, Rockville, MD) was also utilized as a chemokinetic stimulus for peripheral blood T-cells at concentrations of 2 ng/ml, 20 ng/ml, 200 ng/ml and 2 ⁇ g/ml.
  • T-cell subpopulations (1 x 10 5 cells) were mixed with 0.5 ml of 0.5% methylcellulose (Stem Cell Technologies, Vancouver, BC) in sterile medium and plated in a 24 well plate. 5 ⁇ l of PBS or SDF-1 ⁇ at concentrations of 10 ⁇ g/ml or 100ng/ml, was then dispensed gently into 0.5 ml of methylcellulose at a set position on the edge of the well prior to incubation for up to 18 hours on a heated (37°C) microscope stage and the movement of cells viewed by time lapse videomicroscopy.
  • mice C57/B6 mice (Jackson Laboratories, ME) were obtained and primed subcutaneously with 100 ⁇ g chicken ovalbumin (Sigma) containing less than 0.01% LPS by the limulus assay and dissolved in 100 ⁇ l of Complete Freund's Adjuvant (CFA) (Sigma).
  • CFA Complete Freund's Adjuvant
  • Three days ova-primed mice were challenged by intraperitoneal (IP) injection with 100 ⁇ g of ovalbumin (Ova) dissolved in 250 ⁇ l of ddH 2 O. A second 250 ⁇ l IP injection was given to these mice the following day which contained murine SDF-1 ⁇ at 100ng/ml, 1 ⁇ g/ml or 5 ⁇ g/ml.
  • mice were then euthanized 24 hours following the second IP injection and peritoneal lavage with 5 mls of PBS performed with direct visualization of the peritoneal sac and its contents in order to avoid peripheral blood contamination.
  • Peritoneal fluid cells harvested in this way contained ⁇ 0.1% red blood cells.
  • Three groups of control mice were used. In the first, C57/B6 mice were euthanized prior to Ova-priming and a peritoneal lavage performed. The second group of control mice were primed subcutaneously and challenged with Ova intraperitoneally as above and then euthanized 24 hours later. The third group were primed and challenged with Ova as above and after 24 hours injected intraperitoneally with 250 ⁇ l of ddH 2 O alone. These mice were euthanized and a peritoneal lavage performed 24 hours later. Three independent experiments were performed with two or three mice in each experiment.
  • peritoneal fluid harvested from the peritoneal fluid were counted with a hemocytometer and their viability determined by Trypan blue exclusion and the total viable nucleated cell count per ml of peritoneal fluid was determined.
  • Peritoneal fluid harvested in this way was found to contain ⁇ 0.1% red blood cells.
  • Hematological smears of peritoneal fluid were also prepared and stained with Giemsa.
  • Flow cytometry using a FacsCalibur flow cytometer was then performed on peritoneal fluid cells using two panels of anti-murine T-cell monocyte antibodies (anti CD3-PE, anti CD8-PERCP, anti CD4-APC and anti TCR ⁇ , anti-CD11b, anti TCR ⁇ ) (Caltag) and isotype controls.
  • the proportion of T-cells of each subpopulation (CD3 + , CD3 + CD4 + , CD3 + CD8 + ) was determined as a percentage of the total nucleated cell fraction in the peritoneal fluid.
  • SDF-1 ⁇ is a chemokine produced by thymic and bone marrow stroma 12-15 which we evaluated for chemokinetic effects on subpopulations of human T cells, initially within circulating adult T cells and then fetal thymocytes and circulating T cells from the same fetus.
  • Adult peripheral blood T-cells were isolated to a purity of ⁇ 99% by flow cytometry into subpopulations of CD4 + CD45RA + and CD8 + CD45RA + (representing naive cells) and CD4 + CD45RO + and CD8 + CD45RO + (representing memory cells). All populations were represented (15 - 31 % of total lymphoid cells) and demonstrated high percentages of cells positive for the SDF-1 ⁇ receptor, CXCR-4.
  • SDF-1 ⁇ demonstrated chemokinetic and chemoattractant activities for each subpopulation in relative order of CD8 + CD45RA + > CD4 + CD45RA + > CD4 + CD45RO + > CD8 + CD45RO + .
  • Peak chemoattractant activities were seen at a concentration of 100 ng/ml with 97% of CD8 + CD45RA + , 85% of CD4 + CD45RA + , 79% of CD4 + CD45RO + and 51% of CD8 + CD45RO + T-cells transmigrating in response to a positive gradient of SDF-1 ⁇ .
  • the proportion of cells moving away from the SDF-1 ⁇ at 10 ⁇ g/ml was in relative order CD8 + CD45RO + ⁇ CD4 + CD45RA + > CD8 + CD45RA + > CD4 + CD45RO + and was significantly greater than chemokinesis for each subset of cells (p ⁇ 0.01; Student t Test) ( Figure 1A ). Movement of cells away from 10 ⁇ g/ml SDF-1 ⁇ was consistently 2 to 10 times higher than chemokinesis and, for CD8 + CD45RA + and CD8 + CD45RO + cells, greater than chemotaxis.
  • a second cell migration assay was developed in which a tissue culture insert served as a reservoir of SDF-1 ⁇ and was placed in the center of a tissue culture well containing dispersed cells in serum free medium.
  • the insert contained 10 ⁇ g/ml of SDF-1 ⁇
  • adult peripheral blood T-cell subpopulations in the tissue culture well were observed to migrate towards the source of SDF-1 ⁇ and a second population to migrate away. Only movement toward the insert was seen when the insert contained SDF-1 ⁇ at 100 ng/ml.
  • the polarization of T-cell subpopulations both towards and away from SDF-1 ⁇ was also not seen when serum free medium was applied to the insert or when high concentrations of SDF-1 ⁇ existed in both the insert and surrounding medium in the tissue-culture well.
  • T-cell subpopulations (1 x 10 5 cells) were plated in a semi-solid methylcellulose matrix, SDF-1 ⁇ was loaded at a fixed position and cell trafficking was recorded by time lapse videomicroscopy. The movement of a gradient across the methylcellulose was documented in independent experiments by monitoring a dye front through the culture; a process not achieving equilibrium over the duration of our incubations. Consistent movement of peripheral T-cell subpopulations was seen away from high concentrations of SDF-1 ⁇ and towards low concentrations SDF-1 ⁇ .
  • Both CD4 + CD45RA + and CD8 + CD45RA + fetal blood T-cells showed significantly higher levels of chemotaxis and fugetaxis from SDF-1 ⁇ than DP and SP thymocytes derived from the same fetuses (p ⁇ 0.05; Paired t Test).
  • fetal blood T-cells demonstrated maximal chemotactic responses at concentrations of SDF-1 ⁇ of 100 ng/ml whereas fugetaxis was seen at concentrations of SDF-1 ⁇ between 1 ⁇ g and 10 ⁇ g/ml.
  • fetal samples from the same individual demonstrated the acquisition of fugetaxic ability only when T cells had exited the thymus into the circulation.
  • the ability to undergo fugetaxis had marked differentiation stage specificity which was consistent with what we observed in the in vitro T cell differentiation system in which cells moved to the tissue culture well margins when fully differentiated.
  • the tyrosine kinase inhibitors inhibited chemotaxis, but had minimal effect on fugetaxis.
  • the PI-3 kinase inhibitor, wortmannin comparably inhibited fugetaxis, chemotaxis and chemokinesis. This differential sensitivity to kinase inhibitors indicated unique signaling pathways mediating fugetaxis compared with chemotaxis to the same ligand.
  • TSCM also demonstrated clear chemokinetic and chemoattractant activities for each of the four subpopulations of T-cells (see Figure 3 ).
  • the order of chemoattractant activity was CD8+CD45RO+>CD4+CD45RO+> CD4+CD45RA+ >CD8+ CD45RA+.
  • the chemoattractant effect of TSCM could be diluted out and was inhibitable by PTX.
  • TSCM had minimal chemokinetic activities on all the T-cell subpopulations studied.
  • the movement of cells away from TSCM was also demonstrated in all the T-cell subpopulations studied.
  • Chemokines including SDF-1, have been shown to activate multiple signal transduction pathways including phosphatidylinositol 3-kinase (PI-3) and tyrosine kinase ( Ganju, et al., J. Biol. Chem., 1998, 36, 23169 ; Turner, et al., J. Immunol., 1995, 5:2437 ; Turner, et al., J. Biochem. Soc. Trans., 1977, 2:216S ).
  • PI-3 phosphatidylinositol 3-kinase
  • tyrosine kinase Ganju, et al., J. Biol. Chem., 1998, 36, 23169 ; Turner, et al., J. Immunol., 1995, 5:2437 ; Turner, et al., J. Biochem. Soc. Trans., 1977, 2:216S .
  • the signals for fugetaxis or chemotaxis may have
  • the PI-3 kinase inhibitor, wortmannin comparably inhibited fugetaxis, chemotaxis and chemokinesis induced by SDF-1.
  • the tyrosine kinase inhibitors genistein and herbimycin inhibited chemotaxis, but had minimal effect on fugetaxis.
  • Cyclic nucleotides serve as signaling intermediates for multiple G-protein coupled receptors ( Daaka, et al., Nature, 1997, 6655, p88 ; Selbie, et al., Trends Pharmacol Sci, 1998, 3:87-93 ) and have been shown to convert neuronal growth cone responses to netrin-1 from repulsion to attraction ( Song H-J, et al., Science, 1998, 281:1515 ).
  • the membrane-permeable cAMP agonist, 8-Br-cAMP inhibited fugetaxis but not chemotaxis, and that inhibition was concentration dependent. Taken together, these data suggest that fugetaxis and chemotaxis result from the differential activation of signal transduction pathways in response to different concentrations of the same ligand.
  • HIV-1 IIIB gp120 is a bidirectional cue for T-cell migration in vitro
  • HIV-1 gp120 peak concentrations of HIV-1 gp120 of about 200ng/ml caused T-cells to move towards the recombinant retroviral protein. Similar to SDF-1, the bidirectional movement of T-cells induced by HIV-1 IIIB gp120 was inhibited by pertussis toxin and the PI-3 kinase inhibitor wortmannin. In addition, movement away from HIV-1 IIIB gp120 was differentially sensitive to inhibition by the cAMP agonist, 8-Br-cAMP. Furthermore, HIV-1 specific CTL clones derived from HIV-infected patients were shown to exhibit a bidirectional response to X4-HIV-1 gp120.
  • CXCR-4 ligands that include, but are not limited to, SDF-1 ⁇ and ⁇ , met-SDF-1 ⁇ , HIV- 1 IIIB gp120, small molecules T134 and MD3100, T22 [Tyr5,12,Lys7]-polyphemusin II, agonists for such molecules, and the like, have a concentration dependent bifunctional effect on the migration of subpopulations of human T-cells.
  • HepG2CM HepG2 Conditioned Medium
  • HepG2 cell line (derived from a human hepatocarcinoma -ATCC no:HB-8065, ATCC, Manassas, VA) was grown to confluency in MEM + 10% heat inactivated fetal calf serum + penicillin/streptomycin + L-glutamine. Confluent HepG2 cells were then transferred into serum free medium for 48 hours and the serum free conditioned medium (HepG2CM) was harvested from these cultures. HepG2CM was filtered through a 0.4 ⁇ m filter prior to use in transmigration assays or in the Fugetaxin separation system detailed below.
  • Fraction 18 at a 1:10 dilution in serum free medium (original fraction volume between 100-200 ⁇ l), was consistently shown to contain maximal fugetactic activity for Jurkat T-cells as described above.
  • Jurkat T-cells were pretreated with anti-CXCR-4 (Pharmingen) (at 20 ⁇ g/ml for 30 minutes at room temperature), pertussis toxin (PTX) (100 ng/ml for 30 minutes at 37 °C), genistein (1 ⁇ g/ml for 20 minutes at 37 °C), herbimycin (1 ⁇ M for 20 minutes 37 °C), wortmannin (1 ⁇ M for 20 minutes at 37 °C), 8-Br-cAMP (100 ⁇ M for 15 minutes at room temperature) or 8-Br-cGMP (100 ⁇ M for 15 minutes at room temperature) (Sigma Chemical Company, St Louis, MO), prior to their addition to the upper chambers of the transwells in selected experiments.
  • PTX pertussis toxin
  • genistein 1 ⁇ g/ml for 20 minutes at 37 °C
  • herbimycin 1 ⁇ M for 20 minutes 37 °C
  • wortmannin 1 ⁇ M for 20 minutes at 37 °C
  • Transwells prepared in this way were incubated for 3 hours at 37° C /5% CO 2 incubator. Cells were harvested from the lower chamber after 3 hours and cell counts performed. The effect of inhibitors on transmigration of Jurkat cell migration was examined.
  • CXCR-4, genistein and herbamycin inhbited fugetaxis induced by Fraction 18 to 40 - 60% of the control level.
  • PTX inhbited fugetaxis in response to Fraction 18 to 30% of the control level.
  • the cAMP agonist, 8-Br-cAMP resulted in 95% inhbition of fugetaxis whereas 8-Br-cGMP did not significantly inhbit fugetaxis.
  • mice C57 BL/6J mice (Jackson Laboratories, Bar Harbor, ME) were primed subcutaneously with 100 ⁇ g chicken ovalbumin (Ova) (Sigma) containing less than 0.01% LPS by the limulus assay and dissolved in 100 ⁇ l of Complete Freund's Adjuvant (CFA) (Sigma).
  • Ova chicken ovalbumin
  • CFA Complete Freund's Adjuvant
  • IP intraperitoneal
  • mice were euthanized 3 and 24 hours following the second IP injection and peritoneal lavage with 5 mls of PBS performed with direct visualization of the peritoneal sac and its contents in order to avoid peripheral blood contamination.
  • Peritoneal fluid cells harvested in this way contained ⁇ 0.1% red blood cells.
  • mice Three groups of control mice were used. In the first, C57 BL/6J mice were euthanized prior to Ova priming and a peritoneal lavage performed. The second group of control mice were primed subcutaneously and challenged with Ova intraperitoneally as above and then euthanized 3 and 24 hours later. The third group were primed and challenged with Ova as above and injected intraperitoneally with 250 ⁇ l of ddH 2 O alone 24 hours later. These mice were euthanized and a peritoneal lavage performed after a further 3 and 24 hours. Three independent experiments were performed with two or three mice in each group in each experiment.
  • peritoneal fluid harvested from the peritoneal fluid were counted with a hemocytometer and their viability determined by trypan blue exclusion and the total viable nucleated cell count per ml of peritoneal fluid was determined.
  • Peritoneal fluid harvested in this way was found to contain ⁇ 0.1% red blood cells.
  • Hematological smears of peritoneal fluid were also prepared and stained with Giemsa.
  • Flow cytometry using a FACSCalibur flow cytometer was then performed on peritoneal fluid cells using two panels of anti-murine T-cell and monocyte antibodies (anti CD3-PE, anti CD8-Biotin, anti CD4-APC and anti TCR ⁇ , anti-CD11b, anti TCR ⁇ -FITC) (Caltag Laboratories, San Francisco, CA) and isotype controls (Caltag).
  • Second-step staining of biotin-conjugated antibodies was performed using streptavidin-PERCP (Becton Dickinson).
  • the proportion of T-cells of each subpopulation (CD3 + , CD3 + CD4 + , CD3 + CD8 + ) was determined as a percentage of the total nucleated cell fraction in the peritoneal fluid.
  • Fraction 18 was shown to abrogate the T-cell infiltrate induced by IP challenge with Ova. Fraction 22 or pass through did not significantly effect T-cell emigration from the site of antigen challenge ( Figure 6 ).
  • inter- ⁇ -trypsin inhibitor heavy-chain II precursor (ITI H2) family of polypeptides are believed to have similar fugetactic activity, and are therefore useful according to the invention.
  • Such members include the polypeptides having sequences with GenBank Acc. Nos.: IYHU2, NP_034712, NP_002207, Q61703, P97279, O02668, CAA72308 (Y11545), BAA13939 (D89286), S54354, CAA49842(X70392), AAA60558(M18193), CAA30160(X07173), 1409219A, and/or AAA59195(M33033).
  • KSHV Kaposi's sarcoma herpes virus
  • BCBL-1 is a B cell line derived from a body cavity-based B lymphoma that is latently infected with HHV-8/KSHV; no EBV DNA is present in this line ( Renne, R., W. et al., Nat. Med., 1996, 2:342-346 ).
  • T-cell subsets also show a clear chemotactic response to MIP-1 ⁇ at a concentration of 10ng/ml.
  • T-cells demonstrated a clear fugetactic response to ⁇ MIP-I but not ⁇ MIP-II at a concentration of 100ng/ml.

Description

    Field of the Invention
  • This invention relates to methods and compositions for modulating movement of eukaryotic cells with migratory capacity. More specifically, there is described methods and compositions for modulating movement of cells of hematopoietic, neural, epithelial, or mesenchymal origin, in a specific site in a subject. The foregoing are useful, inter alia, in the treatment of conditions characterized by a need to modulate migratory-cell movement associated with specific sites in a subject. Specific sites include sites of inflammation and modulation of migratory-cell movement is movement away from an agent source, or repulsion.
  • Background of the Invention
  • Cell movement in response to specific stimuli is observed to occur in prokaryotes and eukaryotes (Doetsch RN and Seymour WF.,1970; Bailey GB et al.,1985). Cell movement seen in these organisms has been classified into three types; chemotaxis or the movement of cells along a gradient towards an increasing concentration of a chemical; negative chemotaxis which has been defined as the movement down a gradient of a chemical stimulus and chemokinesis or the increased random movement of cells induced by a chemical agent. The receptors and signal transduction pathways for the actions of specific chemotactically active compounds have been extensively defined in prokaryotic cells. Study of E. Coli chemotaxis has revealed that a chemical which attracts the bacteria at some concentrations and conditions may also act as a negative chemotactic chemical or chemorepellent at others (Tsang N et al., 1973; Repaske D and Adler J. 1981; Tisa LS and Adler J.,1995; Taylor BL and Johnson MS., 1998).
  • Chemotaxis and chemokinesis have been observed to occur in mammalian cells (McCutcheon MW, Wartman W and HM Dixon, 1934; Lotz M and H Harris; 1956; Boyden SV 1962) in response to the class of proteins, called chemokines (Ward SG and Westwick J ; 1998; Kim CH et al., 1998; Baggiolini M , 1998; Farber JM; 1997).
  • Chemokines induce cell locomotion by signaling through G-protein coupled receptors (Wells TN et al.,1998). The membrane protein gene superfamily of G-protein coupled receptors has been characterized as having seven putative transmembrane domains. The domains are believed to represent transmembrane α-helices connected by extracellular or cytoplasmic loops. G-protein coupled receptors include a wide range of biologically active receptors, such as hormone, viral, growth factor and neuroreceptors.
  • G-protein coupled receptors have been characterized as including these seven conserved hydrophobic stretches of about 20 to 30 amino acids, connecting at least eight divergent hydrophilic loops. The G-protein family of coupled receptors includes dopamine receptors which bind to neuroleptic drugs used for treating psychotic and neurological disorders. Other examples of members of this family include calcitonin, adrenergic, endothelin, cAMP, adenosine, muscarinic, acetylcholine, serotonin, histamine, thrombin, kinin, follicle stimulating hormone, opsins, endothelial differentiation gene-1 receptor, rhodopsins, odorant, cytomegalovirus receptors, etc.
  • Most G-protein coupled receptors have single conserved cysteine residues in each of the first two extracellular loops which form disulfide bonds that are believed to stabilize functional protein structure. The 7 transmembrane regions are designated as TM1, TM2, TM3, TM4,TM5, TM6, and TM7. TM3 has been implicated in signal transduction. Phosphorylation and lipidation (palmitylation or farnesylation) of cysteine residues can influence signal transduction of some G-protein coupled receptors. Most G-protein coupled receptors contain potential phosphorylation sites within the third cytoplasmic loop and/or the carboxy-terminus. For several G-protein coupled receptors, such as the β-adrenoreceptor, phosphorylation by protein kinase A and/or specific receptor kinases mediates receptor desensitization.
  • For some receptors, the ligand binding sites of G-protein coupled receptors are believed to comprise a hydrophilic socket formed by several G-protein coupled receptors transmembrane domains, which socket is surrounded by hydrophobic residues of the G-protein coupled receptors. The hydrophilic side of each G-protein coupled receptor transmembrane helix is postulated to face inward and form the polar ligand binding site. TM3 has been implicated in several G-protein coupled receptors as having a ligand binding site, such as including the TM3 aspartate residue. Additionally, TM5 serines, a TM6 asparagine and TM6 or TM7 phenylalanines or tyrosines are also implicated in ligand binding.
  • G-protein coupled receptors can be intracellularly coupled by heterotrimeric G-proteins to various intracellular enzymes, ion channels and transporters (see, Johnson et al., Endoc Rev, 1989, 10:317-331). Different G-protein a-subunits preferentially stimulate particular effectors to modulate various biological functions in a cell. Phosphorylation of cytoplasmic residues of G-protein coupled receptors have been identified as an important mechanism for the regulation of G-protein coupling of some G-protein coupled receptors. G-protein coupled receptors are found in numerous sites within a mammalian host. Chemokine-induced cell chemotaxis and chemokinesis are thought to be mediated via a Gαi-linked signal transduction pathway and can be blocked by pertussis toxin (PTX) (Luster AD, 1998; Baggiolini, 1998).
  • The chemokine, SDF-1α, causes immigration of subpopulations of leukocytes into sites of inflammation (Aiuti A et al. 1997; Bleul CC et al. 1996; Bleul CC et al., 1996; Oberlin E et al., 1996). Furthermore, mice engineered to be deficient in SDF-1α or its receptor, CXCR-4, have abnormal development of hematopoietic tissues and B-cells manifesting a failure of fetal liver stem cells to migrate to bone marrow (Friedland JS, 1995; Tan J and Thestrup-Pedersen K, 1995; Corrigan CJ and Kay AB, 1996; Qing M, et al,1998; Ward SG et al. 1998).
  • Although chemotaxis and chemokinesis have been defined in cell subpopulations in mammals, negative chemotaxis of peripheral blood cells from mammals has only been observed in response to non-specific stimuli such as cell lysates or tumor tissue fragments (Jochims, 1927; McCutcheon et al. 1939; Bessis M and Burte B., 1965; Noble and Bentley, 1981) and reports have been very limited. The precise mechanism of negative chemotaxis in higher eukaryotic cell subpopulations, including the definition of specific stimuli and signal transduction pathways of negative chemotaxis has not been defined. Furthermore, although chemoattractants had been shown to serve as repellents in prokaryotic systems, no analogous system of a dual action chemoattractant/chemorepellent compound has been identified in higher eukaryotes.
  • Summary of the Invention
  • We describe herein the isolation of agents with migratory-cell repellant activity (hereinafter "fugetactic agents" and "fugetactic activity," and/or "chemo-fugetaxis"). We also describe the identification of previously isolated agents as fugetactic agents at defined concentrations. There is described pharmaceutical compositions containing the foregoing fugetactic agents, and various therapeutic and diagnostic methods utilizing the foregoing fugetactic agents. There is also described isolated fugetactic polypeptides and agents which bind such polypeptides, including antibodies. The foregoing can be used, inter alia, in the treatment of conditions characterized by a need to modulate migratory-cell movement in specific sites in a subject. Important such sites include inflammation sites. There is also described methods for identifying agents useful in the modulation of such fugetactic activity.
  • According to a first aspect of the present invention there is provided use of a fugetactic agent in the manufacture of a medicament for treating inflammation, autoimmune disease, an abscess, a transplant, an implant, atherosclerosis, myocarditis, infertility, or a tumor, the medicament containing a fugetactic amount of said agent for repelling immune cells from a specific site in a subject when the medicament is locally administered to the specific site in the subject, wherein the fugetactic agent is an agent having migratory cell repellent activity on a cell in a fugetactic amount, and having chemoattractant activity on the same cell at lower concentrations, wherein the fugetactic agent is SDF-1α or HIV-IIIIgp120.
  • According to another aspect of the present invention there is provided use of a fugetactic agent in the manufacture of a medicament containing a fugetactic amount of said agent for repelling immune cells away from a specific site in a subject, wherein the fugetactic agent is SDF-1α or HIV-IIIBgp120.
  • According to another aspect of the present invention there is provided a method for distinguishing mature T cells from immature T cells in vitro, comprising: contacting a fugetactic agent with a mixed population of mature and immature T cells and measuring cell movement relative to the fugetactic agent, wherein movement of the cells away from the fugetactic agent is indicative of mature T cells; wherein the fugetactic agent is SDF-1α or HIV=IIIIBgp120.
  • According to another aspect of the present invention there is provided a medicament containing a fugetactic agent for use in treating inflammation, autoimmune disease, an abscess, a transplant, an implant, atherosclerosis, myocarditis, infertility, or a tumor, wherein the medicament contains a fugetactic amount of said agent for repelling immune cells from a specific site in a subject when the medicament is locally administered to the specific site in the subject, wherein the fugetactic agent is an agent having migratory cell repellent activity on a cell in a fugetactic amount, and having chemoattractant activity on the same cell at lower concentrations, wherein the fugetactic agent is SDF-1α or HIV-IIIIBgp120.
  • According to another aspect of the present invention there is provided use of a fugetactic agent to coat a material surface of an implant wherein the fugetactic agent is present in amount effective to repel immune cells from the material surface, wherein the fugetactic agent is SDF-1α or HIV-IIIIBgp120.
  • According to another aspect of the present invention there is provided an implant suitable for implanting into a subject wherein the implant comprises a material surface coated with an amount of a fugetactic agent effective to repel immune calls from the material surface, wherein the fugetactic agent is SDF-1α or HIV-IIIIBgp120.
  • There is described herein a method of inhibiting migration of immune cells to a specific site in a subject. The method involves locally administering to a specific site in a subject in need of such treatment a fugetactic agent in an amount effective to inhibit migration of immune cells to the specific site in a subject. The specific site may be a site of inflammation. When the specific site is the site of inflammation, the method may further comprise co-administering a non-fugetactic agent that inhibits migration of immune cells to the site of inflammation in the subject. The non-fugetactic agent may include an anti-inflammatory agent and/or an immunosuppressant.
  • The subject may have an autoimmune disease. The autoimmune disease may include rheumatoid arthritis, uveitis, insulin-dependent diabetes mellitus, hemolytic anemias, rheumatic fever, Crohn's disease, Guillain-Barre syndrome, psoriasis, thyroiditis, Graves' disease, myasthenia gravis, glomerulonephritis, autoimmune hepatitis, systemic lupus erythematosus. In further embodiments, the subject has multiple sclerosis, an abscess, a transplant, an implant, atherosclerosis, and/or myocarditis.
  • In any of the foregoing aspects and embodiments of the invention, the fugetactic agent is
    HIV-IIIIB gp120 or SDF-1α.
  • In some embodiments, the fugetactic agent is SDF-1α at a concentration higher than about 1µg/ml or HIV-1IIIB gp120 at a concentration higher than about 200ng/mL
  • There is described herein a method for promoting migration of cells away from a specific site in a subject. The method involves locally-administering to a specific site in a subject in need of such treatment a fugetactic agent in an amount effective to promote migration of cells away from the specific site in the subject. The specific site may be an inflammation site. The specific site may be a germ cell-containing site. The specific site may be an area surrounding a tumor, but not the immediate area surrounding the tumor. The specific site may be a transplanted tissue and/or an implant. Important fugetactic agents, cell-types, and so on, are as described above. In any of the foregoing description, preferred cells are immune cells.
  • According to a further aspect of the invention, a method for distinguishing mature T-cells from immature T-cells in vitro is provided. The method involves contacting SDF-1 or HIViiibgp120 with a mixed population of mature and immature T-cells and measuring cell movement relative to the fugetactic agent, wherein movement of cells away from the fugetactic agent is indicative of mature T-cells.
  • There is also described herein a method of screening for fugetactic agents. The method involves contacting an agent suspected of being a fugetactic agent with a cell with migratory capacity, measuring movement of the cell with migratory capacity relative to the agent, and determining whether the movement of the cell with migratory capacity is movement away from the agent, wherein movement of the cell with migratory capacity away from the agent is indicative of the agent being a fugetactic agent. The cell with migratory capacity may be a hematopoietic cell. The cell with migratory capacity may be a neural cell. The cell with migratory capacity may be an epithelial cell. The cell with migratory capacity may be a mesenchymal cell. The cell with migratory capacity may be an embryonic stem cell. The cell with migratory capacity may be a germ cell. The agent depicted of being a fugetactic agent may be known to be a chemoattractant at a defined concentration and a fugetactic agent at a concentration higher than the defined concentration. Such fugetactic agents may include cytokines. The agent suspected of being a fugetactic agent may be an agent present in a biological fluid. The biological fluid may include synovial fluid, cerebral spinal fluid, fellopian tube fluid, seminal fluid, ocular fluid, pericardial fluid, pleural fluid, inflammatory exudate and ascitic fluid. The agent suspected of being a fugetactic agent may be an agent present in a tumor cell culture supernatant, tumor cell eluate and/or tumor cell lysate. The tumor cells may be HepG2 cells and the cell with migratory capacity may be a hematopoietic cell. The agent suspected of being a fugetactic agent may be an expression product of a cDNA library.
  • There is also described herein a method of repelling immune cells from a material surface. The method involves coating a material surface with an amount of a fugetactic agent effective to repel immune cells from the material surface. The material surface may be part of an implant. The material comprising the implant may be synthetic material or organic tissue material. Important fugetactic agents, cell-types, and so on, are as described above.
  • There is also described herein a method of screening for an agent that modulates migratory cell-specific fugetactic activity of a fugetactic agent. The method involves forming a mixture comprising a cell with migratory capacity, a fugetactic agent and a candidate anti-fugetactic agent incubating the mixture under conditions which permit binding of the candidate anti-fugetactic agent to the fugetactic agent, determining the level of a test fugetactic activity of the fugetactic agent and the candidate anti-fugetactic agent on the cells with migratory capacity, and comparing the level of the test fugetactic activity to a control level of fugetactic activity determined in the absence of the candidate anti-fugetactic agent. Reduction in the test fugetactic activity level relative to the control level of fugetactic activity may indicate that the candidate anti-fugetactic agent is a lead compound for an anti-fugetactic agent which inhibits the fugetactic activity of the fugetactic agent. An increase in the test fugetactic activity level relative to control level of fugetactic activity may indicate that the candidate anti-fugetactic agent is a lead compound for an anti-fugetactic agent which increases the fugetactic activity of the fugetactic agent. The fugetactic agent may be SDF-1α at a concentration higher than about 1µg/ml or HIV-1IIIB gp120 at a concentration higher than about 200ng/ml.
  • The cell with migratory capacity may be a hematopoietic cell, a neural cell, an epithelial cell, a mesenchymal cell, an embryonic stem cell, a cell involved in angiogenesis (blood vessel formation), or a germ cell.
  • There is also described herein a method of enhancing an immune response in a subject having a condition that involves a specific site. The method involves locally administering to a specific site in a subject in need of such treatment an anti-fugetactic agent in an amount effective to inhibit immune cell-specific fugetactic activity at the specific site in the subject. The specific site may be a site of a pathogenic infection. The specific site may be a germ cell-containing site.
  • The specific site may be an area immediately surrounding a tumor. The anti-fugetactic agent described herein may be a cytokine binding agent such as an anti-cytokine antibody or a cytokine agonist. The cytokine may be SDF-1α.
  • There is also described herein a method of inhibiting tumor cell metastasis in a subject. The method involves locally administering to a tumor site in a subject in need of such treatment an anti-fugetactic agent in an amount effective to inhibit metastasis of tumor cells from the tumor site in the subject. The anti-fugetactic agent may be as described above.
  • There is also described herein a method of inhibiting endothelial cell migration to a tumor site in a subject. The method involves locally administering to an area surrounding a tumor site in a subject in need of such treatment a fugetactic agent in an amount effective to inhibit endothelial cell migration to the tumor site in the subject. The area surrounding the tumor site may not be immediate to the tumor site. Important fugetactic agents are as described above.
  • There is also described herein a method of contraception in a subject. The method involves administering to a subject in need of such treatment, an anti- fugetactic agent in an amount effective to inhibit germ cell migration in the subject. The anti-fugetactic agent may be as described above.
  • There is also described herein a method of treating infertility and premature labor, including premature delivery and impending miscarriage. The method involves administering to a subject in need of such treatment a fugetactic agent in an amount effective to inhibit immune cells from migrating close to a germ cell in the subject. The administration may be local to a germ cell-containing site of the subject.
  • In any of the foregoing aspects and embodiments of the invention where the fugetactic agent is identified as SDF-1α. or HIV-IIIIB gp120, fragments of such polypeptide useful according to the invention are only those with fugetactic activity. A person of ordinary skill in the art could easily determine which fragments have such activity by first creating deletions of the full length polypeptides using methods well known in the art, and testing such fragments for their fugetactic activity according to the teachings of the present invention (see, e.g., Examples).
  • These and other aspects of the invention, as well as various advantages and utilities, will be more apparent with reference to the detailed description of the preferred embodiments.
  • Brief Description of the Drawings
    • Figure 1(A) . Bar graph showing that SDF-1α induces fugetaxis, chemotaxis and chemokinesis in various T-cell subsets; Figure 1(B) . Graphs showing the effects of various SDF-1α concentrations on T-cell subsets.
    • Figure 2 . Dose response curves to SDF-1α, showing that both fugetaxis and chemotaxis are inhibitable by pertussis toxin.
    • Figure 3 . Bar graph displaying the effect of human thymic stromal-cell isolate (TSCM) on peripheral blood T cell populations.
    • Figure 4 . Graphs showing that CD8+CD45RA+ T-cells fugetax in a CD4-independent, CXCR-4 dependent (Fig. 12B), concentration dependent manner (Figs. 12A and 12C) in response to recombinant HIV-IIIIB gp120.
    • Figure 5 . Graphs depicting that SDF-1 at high concentration (5µg/ml), causes T cell migration away from a site of secondary immune response in vivo.
    • Figure 6 . Graph depicting that HepG2CM Fraction 18 abrogates T-cell infiltration to a site of antigenic challenge in vivo.
    • Figure 7 . Graphs showing that T-cell subsets fugetax in response to KSHV-infected cell-isolates (supernatants); (A) BCBL-1 cells; (B) VG-1 cells.
    Brief Description of the Sequences
  • SEQ ID NO: 1 is the amino acid sequence of inter-α-trypsin inhibitor heavy-chain II precursor (ITI H2) (GenBank Acc. No. P 19823).
  • Detailed Description of the Invention
  • There is described herein the discovery of agents with eukaryotic migratory-cell repellant activity (hereinafter "fugetactic agents" and "fugetactic activity"). The invention involves the unexpected discovery of agents, previously known to be chemoattractants at defined concentrations, to be acting as fugetactic agents at concentrations higher than the defined concentrations. Pharmaceutical compositions containing the foregoing fugetactic agents, and various therapeutic and diagnostic methods utilizing the foregoing fugetactic agents, are also described in more detail below. The foregoing can be used, inter alia, in the treatment of conditions characterized by a need to modulate migratory-cell movement in specific sites in a subject.
  • As used herein, a subject is a human, non-human primate, cow, horse, pig, sheep, goat, dog, cat or rodent. In all embodiments, human subjects are preferred.
  • By "fugetactic activity" it is meant the ability of an agent to repel (or chemorepel) a eukaryotic cell with migratory capacity (i.e., a cell that can move away from a repellant stimulus). Accordingly, an agent with fugetactic activity is a "fugetactic agent." Such activity can be detected using any of the transmigration systems described herein (see Examples), or a variety of other systems well known in the art (see, e.g., U.S. patent 5,514,555, entitled: "Assays and therapeutic methods based on lymphocyte chemoattractants," issued May 7, 1996, to Springer, TA, et al. ).
  • There is described herein a thymic stromal-cell isolate with fugetactic activity, repelling cells with migratory capacity and in particular mature T cells. Therefore, the thymic stromal-cell isolate is useful generally in conditions where inhibition of migration of immune cells to a specific site is desirable. Such conditions are described in greater detail below and in their majority are associated with an inflammatory response. As described in greater detail below, the thymic stromal-cell isolate is produced by thymic stromal cells. The thymic stromal-cell isolate is protease and heat sensitive, and it elutes-off a typical sepharose ion exchange column with 0.5M NaCl at pH 6.3, and wherein the agent is not SDF-1α. The thymic stromal-cell isolate may comprise a polypeptide, and it is this polypeptide that repels immune cells. The thymic stromal-cell isolate does not contain an immune cell repelling concentration of SDF-1α. An "immune cell repelling concentration of SDF-1α," as used herein, is any concentration of SDF-1α higher than about 100ng/ml.
  • An immune cell repelling concentration of SDF-la may also be any concentration of SDF-1α higher than about 150ng/ml. An immune cell repelling concentration of SDF-1α may also be any concentration of SDF-1α higher than a concentration selected from the group consisting of about 300ng/ml, 500ng/ml, 750ng/ml, and 1µg/ml.
  • As used herein with respect to thymic stromal-cell isolate, "isolate (i.e. isolated)" means separated from its native environment and present in sufficient quantity to permit its identification or use. Isolated, when referring to a protein or polypeptide, means, for example: (i) selectively produced by expression cloning or (ii) partially purified as by chromatography or electrophoresis. Isolated proteins or polypeptides may, but need not be, substantially pure. The thymic stromal-cell isolate may be substantially pure polypeptide. The term "substantially pure" means that the protein(s) or polypeptide(s) is essentially free of other substances with which it (they) may be found in nature or in vitro systems, to an extent practical and appropriate for their intended use. Substantially pure polypeptides may be produced by techniques well known in the art. Because an isolated protein may be admixed with a pharmaceutically acceptable carrier in a pharmaceutical preparation, the protein may comprise only a small percentage by weight of the preparation. The protein is nonetheless isolated in that it has been separated from many of the substances with which it may be associated in living systems, i.e. isolated from certain other proteins.
  • The fugetactic polypeptide (or thymic stromal-cell isolate) can be isolated from a non-homogenous proteinaceous solution such as a cell culture supernatant or cell homogenate. Thymic stromal-cells can be isolated from a subject by the disaggregation of a piece of thymic tissue, and forming cell suspensions. These thymic stromal-cell suspensions can be cultured according to standard cell culture techniques. In small scale, the cultures can be contained in culture plates, flasks, and dishes. In larger scale, the cultures can be contained in roller bottles, spinner flasks and other large scale culture vessels such as fermenters. Culturing in a three-dimensional, porous, solid matrix may also be used. In preferred embodiments, thymic stromal-cells are obtained from human fetal thymi of 16 to 22 week abortuses as described in greater detail below.
  • Conveniently, the thymic stromal-cell isolate can be isolated from the supernatants of the above-described cell cultures, although the entire culture can be homogenized and subjected to the steps described below for isolation of a fugetactic polypeptide (thymic stromal-cell isolate). Typically the supernatant is removed by aspiration or by centrifugation of the cell culture to remove the cells. The cultures can also be filtered to remove cells and cell debris. In important embodiments, the collected supernatant is (in its entirety) the thymic stromal-cell isolate.
  • The fugetactic polypeptide (thymic stromal-cell isolate)-containing supernatant can be fractionated according to standard chromatographic procedures to facilitate isolation of the fugetactic polypeptide (or thymic stromal-cell isolate). One of ordinary skill in the art will be familiar with such procedures that include, but are not limited to, size-exclusion chromatography, FPLC, HPLC, gel filtration chromatography, ion-exchange chromatography, hydrophobic chromatography, immune-affinity chromatography, etc.
  • The fractions of thymic stromal-cell isolate containing supernatant may be used to repel immune cells, particularly mature T cells. "Immune cells" as used herein are cells of hematopoietic origin (see later discussion), that are involved in the specific recognition of antigens. Immune cells include antigen presenting cells (APCs), such as dendritic cells or macrophages, B cells, T cells, etc. "Mature T cells" as used herein include T cells of a CD4loCD8hiCD69+TCR+, CD4hiCD8loCD69+TCR+, CD4+CD3+ RO+ and/or CD8+CD3+ RO+ phenotype.
  • The fugetactic response of the mature T cells to the thymic stromal-cell isolate can be measured as described above, or according to the transmigration assays described in greater detail in the Examples. Other suitable methods will be known to one of ordinary skill in the art and can be employed using only routine experimentation.
  • The fractions which are positive for the thymic stromal-cell isolate can be subjected to additional rounds of screening using the foregoing methodology. The purity of the fraction can be assessed after each round of culture stimulation by subjecting an aliquot of the fraction to SDS-PAGE or other analytical method for visualizing the mixture of constituents in the fraction. The nature of the thymic stromal-cell isolate as a protein, nucleic acid, lipid, carbohydrate etc., can be confirmed at any time by treating an aliquot of a positive fraction with non-specific degradative enzymes for the foregoing classes of molecules and testing the treated fraction in the same assays detailed above.
  • The thymic stromal-cell isolate can then be further isolated if desired using immunological and molecular biological methods (see, e.g. Molecular Cloning: A Laboratory Manual, J. Sambrook, et al., eds., Second Edition, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 1989, or Current Protocols in Molecular Biology, F.M. Ausubel, et al., eds., John Wiley & Sons, Inc., New York). For example, a fraction positive for the thymic stromal-cell isolate (i.e. with fugetactic activity) which is sufficiently purified can be subjected to protein sequencing according to standard methods. For example, the fraction can be subjected to SDS-PAGE, transferred to a membrane such as polyvinylidene fluoride by electroblotting, and N-terminal amino sequence determined by Edman degradation. Any sequence information can be used to screen databases for homology to existing proteins and also to generate degenerate nucleic acids useful for screening a cDNA library by standard methods such as colony hybridization or polymerase chain reaction. Alternatively, the positive fraction can be used to generate antibodies which recognize the thymic stromal-cell isolate. Such antibodies can then be used in expression cloning protocols, Western blots, and other techniques useful in isolation of the thymic stromal-cell isolate. In the foregoing methods, any cDNA libraries, expression libraries, etc., are preferably created from thymic stromal-cells.
  • The description also makes it possible to isolate proteins which bind to the thymic stromal-cell isolate as disclosed herein, including antibodies and cellular binding partners of the thymic stromal-cell isolate such as receptors. Once the thymic stromal-cell isolate is isolated according to standard methods known to one of ordinary skill in the art, the thymic stromal-cell isolate (or even a substantially purified cell supernatant or fraction) can be used to generate polyclonal or monoclonal antibodies according to standard methods (see e.g., Harlow and Lane, eds., Antibodies: A Laboratory Manual, Cold Spring Harbor Press, Cold Spring Harbor, NY, 1988)
  • The proteins which bind to the thymic stromal-cell isolate can be used, for example, in screening assays to detect the presence or absence of thymic stromal-cell isolate and complexes of thymic stromal-cell isolate and their respective binding partners and in purification protocols to isolate thymic stromal-cell isolate and complexes of thymic stromal-cell isolate and their respective binding partners. The binding proteins also can be used to block the effects of the thymic stromal-cell isolate. Such assays can be used to confirm the specificity of binding.
  • The present disclosure therefore, embraces peptide binding agents which, for example, can be antibodies or fragments of antibodies having the ability to selectively bind to thymic stromal-cell isolate and inhibit the fugetactic properties of the isolate or agent (or agent as described above). The peptide binding agents may bind selectively to a substantially pure organic agent that repels mature T-cells. The peptide binding agents may bind selectively to SDF-1α and inhibit repulsion of immune cells. Antibodies include polyclonal and monoclonal antibodies, prepared according to conventional methodology.
  • Significantly, as is well-known in the art, only a small portion of an antibody molecule, the paratope, is involved in the binding of the antibody to its epitope (see, in general, Clark, W.R. (1986) The Experimental Foundations of Modern Immunology Wiley & Sons, Inc., New York; Roitt, I. (1991) Essential Immunology, 7th Ed., Blackwell Scientific Publications, Oxford). The pFc' and Fc regions, for example, are effectors of the complement cascade but are not involved in antigen binding. An antibody from which the pFc' region has been enzymatically cleaved, or which has been produced without the pFc' region, designated an F(ab')2 fragment, retains both of the antigen binding sites of an intact antibody. Similarly, an antibody from which the Fc region has been enzymatically cleaved, or which has been produced without the Fc region, designated an Fab fragment, retains one of the antigen binding sites of an intact antibody molecule. Proceeding further, Fab fragments consist of a covalently bound antibody light chain and a portion of the antibody heavy chain denoted Fd. The Fd fragments are the major determinant of antibody specificity (a single Fd fragment may be associated with up to ten different light chains without altering antibody specificity) and Fd fragments retain epitope-binding ability in isolation.
  • Within the antigen-binding portion of an antibody, as is well-known in the art, there are complementarity determining regions (CDRs), which directly interact with the epitope of the antigen, and framework regions (FRs), which maintain the tertiary structure of the paratope (see, in general, Clark, 1986; Roitt, 1991). In both the heavy chain Fd fragment and the light chain of IgG immunoglobulins, there are four framework regions (FR1 through FR4) separated respectively by three complementarity determining regions (CDR1 through CDR3). The CDRs, and in particular the CDR3 regions, and more particularly the heavy chain CDR3, are largely responsible for antibody specificity.
  • It is now well-established in the art that the non-CDR regions of a mammalian antibody may be replaced with similar regions of conspecific or heterospecific antibodies while retaining the epitopic specificity of the original antibody. This is most clearly manifested in the development and use of "humanized" antibodies in which non-human CDRs are covalently joined to human FR and/or Fc/pFc' regions to produce a functional antibody. Thus, for example, PCT International Publication Number WO 92/04381 teaches the production and use of humanized murine RSV antibodies in which at least a portion of the murine FR regions have been replaced by FR regions of human origin. Such antibodies, including fragments of intact antibodies with antigen-binding ability, are often referred to as "chimeric" antibodies.
  • Thus, as will be apparent to one of ordinary skill in the art, the present invention also provides for F(ab')2, Fab, Fv and Fd fragments; chimeric antibodies in which the Fc and/or FR and/or CDR1 and/or CDR2 and/or light chain CDR3 regions have been replaced by homologous human or non-human sequences; chimeric F(ab')2 fragment antibodies in which the FR and/or CDR1 and/or CDR2 and/or light chain CDR3 regions have been replaced by homologous human or non-human sequences; chimeric Fab fragment antibodies in which the FR and/or CDR1 and/or CDR2 and/or light chain CDR3 regions have been replaced by homologous human or non-human sequences; and chimeric Fd fragment antibodies in which the FR and/or CDR1 and/or CDR2 regions have been replaced by homologous human or non-human sequences. The present invention also includes so-called single chain antibodies.
  • There is also described herein polypeptides of numerous size and type that bind specifically to thymic stromal-cell isolate, and complexes of both thymic stromal-cell isolate and its binding partners. These polypeptides may be derived also from sources other than antibody technology. For example, such polypeptide binding agents can be provided by degenerate peptide libraries which can be readily prepared in solution, in immobilized form or as phage display libraries. Combinatorial libraries also can be synthesized of peptides containing one or more amino acids. Libraries further can be synthesized of peptoids and non-peptide synthetic moieties.
  • As detailed herein, the foregoing antibodies and other binding molecules may be used for example to identify tissues expressing protein or to purify protein. Antibodies also may be coupled to specific diagnostic labeling agents for imaging of cells and tissues that express a thymic stromal-cell isolate.
  • The invention relates to the unexpected discovery that agents previously known to be acting as chemoattractants of immune cells at defined concentrations, also act as fugetactic agents to the same cells at concentrations higher than the defined concentrations.
  • Such fugetactic agents include cytokines. "Cytokine" is a generic term for nonantibody soluble proteins which are released from one cell subpopulation and which act as intercellular mediators, for example, in the generation or regulation of an immune response. See Human Cytokines: Handbook for Basic & Clinical Research (Aggrawal, et al. eds., Blackwell Scientific, Boston, Mass. 1991). Cytokines include, e.g., interleukins IL-1 through IL-15, tumor necrosis factors α & β, interferons α, β, and γ, tumor growth factor beta (TGF-β), colony stimulating factor (CSF) and granulocyte monocyte colony stimulating factor (GM-CSF). The action of each cytokine on its target cell is mediated through binding to a cell surface receptor. Cytokines share many properties of hormones, but are distinct from classical hormones in that in vivo, they generally act locally on neighboring cells within a tissue. The activities of cytokines range from promoting cell growth (e.g., IL-2, IL-4, and IL-7), and arresting growth (IL-10, tumor necrosis factor and TGF-β), to inducing viral resistance (IFN α, β, and γ). See Fundamental Immunology (Paul ed., Raven Press, 2nd ed. 1989); Encyclopedia of Immunology, (Roitt ed., Academic Press 1992) (which are hereby incorporated by reference in their entirety for all purposes). In certain embodiments, the cytokine is a cytokine with chemoattractant and/or chemokinetic properties. Examples of such cytokines include: PAF, N-formylated peptides, C5a, LTB4, LXA4, chemokines: CXC, IL-8, GCP-2, GROα, GROβ, GROγ, ENA-78, NAP-2, IP-10, MIG, I-TAC, SDF-1α, BCA-1, PF4, Bolekine, MIP-1α, MIP-1β, RANTES, HCC-1, MCP-1, MCP-2, MCP-3, MCP-4, MCP-5 (mouse only), Leukotactin-1 (HCC-2, MIP-5), Eotaxin, Eotaxin-2 (MPIF2), Eotaxin-3 (TSC), MDC, TARC, SLC (Exodus-2, 6CKine), MIP-3α (LARC, Exodus-1), ELC (MIP-3β), I-309, DC-CK1 (PARC, AMAC-1), TECK, CTAK, MPIF1 (MIP-3), MIP-5 (HCC-2), HCC-4 (NCC-4), MIP-1γ (mouse only), C-10 (mouse only); C: Lymphotactin; CX3C: Fracktelkine (Neurotactin). Cytokines may be members of the Cys-X-Cys family of chemokines (chemokines that bind to the CXCR-4 receptor). Such agents include SDF-1α, SDF-1β, and met- SDF-1 β. The cytokine used in the present invention is SDF-1α. Fugetactic agents include other CXCR-4 receptor ligands CXCR-4 ligands include, but are not limited to, HIV-IIIIB gp120, small molecules T134 and MD3100, and/or T22 ([Tyr5,12,Lys7]-polyphemusin II) (Heveker et al., Curr Biol, 1998, 8:369-76). The CXCR-4 ligand used in the present invention is HIV-IIIIBgp120.
  • SDF-1α is a cytokine (chemokine) produced by thymic and bone marrow stroma (12-15 and U.S. Patent 5,756,084, entitled: "Human stromal derived factor 1α and 1β," issued May 26, 1998, to Honjo, et al. ), that has been reported as a highly efficacious and highly potent lymphocyte chemoattractant at concentrations lower than about 100ng/ml. We have discovered, unexpectedly, that SDF-1α at a concentration higher than about 200ng/ml, and preferably higher than about 1 µg/ml, acts a potent fugetactic agent to immune cells, and more specifically to mature T cells, as described in greater detail in the Examples.
  • Recombinant envelope glycoprotein gp120 of substrain IIIB of HIVLAI (HIV-1IIIB gp120) has been used extensively in the art to generate neutralizing antibodies to HIV. The association between gp120 and the CXCR-4 co-receptor for efficient HIV entry into a cell has also been established (Stott EJ, et al., J Gen Virol, 1998, 79(Pt3):423-32). We have discovered, that HIV-1IIIB gp120 at a concentration higher than about 10ng/ml, and preferably higher than about 200ng/ml, acts a potent fugetactic agent to immune cells, and more specifically to mature T cells, as described in greater detail in the Examples.
  • There is also described herein a method for isolating a stromal-cell derived fugetactic agent. According to the method, a culture of stromal-cells is first prepared. Stromal cells comprising fibroblasts with or without other cells and elements described below (the term "fibroblasts" is used collectively herein to include such cells), are inoculated onto a tissue culture vessel (a typical two-dimensional culture dish or three-dimensional matrix). These fibroblasts may be derived from tissues or organs that include, but are not limited to, skin, liver, pancreas, bone marrow, lymph node, thymus, kidney, CNS, brain, etc., which can be obtained by biopsy (where appropriate) or upon autopsy. In fact fibroblasts can be obtained in quantity rather conveniently from any appropriate cadaver organ. Fetal fibroblasts can also be used.
  • Fibroblasts may be readily isolated by disaggregating an appropriate organ or tissue which is to serve as the source of the fibroblasts. This may be readily accomplished using techniques known to those skilled in the art. For example, the tissue or organ can be disaggregated mechanically and/or treated with digestive enzymes and/or chelating agents that weaken the connections between neighboring cells, making it possible to disperse the tissue into a suspension of individual cells without appreciable cell breakage. Enzymatic dissociation can be accomplished by mincing the tissue and treating the minced tissue with any of a number of digestive enzymes either alone or in combination. These include but are not limited to trypsin, chymotrypsin, collagenase, elastase, and/or hyaluronidase, DNase, pronase, dispase, etc. Mechanical disruption can also be accomplished by a number of methods including, but not limited to, the use of grinders, blenders, sieves, homogenizers, pressure cells, or insonators to name but a few. For a review of tissue disaggregation techniques, see Freshney, Culture of Animal Cells, A Manual of Basic Techniques, 2d Ed., A. R. Liss, Inc., New York, 1987, Ch. 9, pp. 107-126.
  • Once the tissue has been reduced to a suspension of individual cells, the suspension can be fractionated into subpopulations from which the fibroblasts and/or other stromal cells and/or elements can be obtained. This also may be accomplished using standard techniques for cell separation including, but not limited to, cloning and selection of specific cell types, selective destruction of unwanted cells (negative selection), separation based upon differential cell agglutinability in the mixed population, freeze-thaw procedures, differential adherence properties of the cells in the mixed population, filtration, conventional and zonal centrifugation, centrifugal elutriation (counter-streaming centrifugation), unit gravity separation, countercurrent distribution, electrophoresis and fluorescence-activated cell sorting. For a review of clonal selection and cell separation techniques, see Freshney, Culture of Animal Cells, A Manual of Basic Techniques, 2d Ed., A. R. Liss, Inc., New York, 1987, Ch. 11 and 12, pp. 137-168.
  • The isolation of fibroblasts may, for example, be carried out as follows: fresh tissue samples are thoroughly washed and minced in Hank's balanced salt solution (HBSS) in order to remove serum. The minced tissue is incubated from 1-12 hours in a freshly prepared solution of a dissociating enzyme such as trypsin. After such incubation, the dissociated cells are suspended, pelleted by centrifugation and plated onto culture dishes. All fibroblasts will attach before other cells, therefore, appropriate stromal cells can be selectively isolated and grown. The isolated fibroblasts can then be grown to confluency, lifted from the confluent culture and inoculated onto the three-dimensional matrix (see, Naughton et al., 1987, J. Med., 18:219-250). Inoculation of the culture vessel with a high concentration of stromal cells, e.g., approximately 106 to 5 x 107 cells/ml, will result in the establishment of a confluent stromal-cell culture in shorter periods of time.
  • As mentioned above, other stromal-cells found in loose connective tissue may be inoculated onto the culture vessel along with the fibroblasts. These stromal-cells may readily be derived from the same appropriate organs the fibroblasts are derived, that include, but are not limited to, skin, liver, pancreas, bone marrow, lymph node, thymus, kidney, CNS, brain, etc., using methods known in the art such as those discussed above. Such cells include, but are not limited to, endothelial cells, pericytes, macrophages, monocytes, plasma cells, mast cells, adipocytes, etc. In one example, stromal cells of hematopoietic tissue, including but not limited to, fibroblasts, endothelial cells, macrophages/monocytes, adipocytes and reticular cells, could be used to inoculate a tissue culture dish and/or a three-dimensional matrix.
  • Hematopoietic stromal-cells may be readily obtained from the "buffy coat" formed in bone marrow suspensions by centrifugation at low forces, e.g., 3000g. Stromal-cells of liver may include fibroblasts, Kupffer cells, and vascular and bile duct endothelial cells. Similarly, glial cells could be used as the stroma to support the proliferation of neurological cells and tissues; glial cells for this purpose can be obtained by trypsinization or collagenase digestion of embryonic or adult brain (Ponten and Westermark, 1980, in Federof, S. Hertz, L., eds, Advances in Cellular Neurobiology, Vol. 1, New York, Academic Press, pp.209-227).
  • After inoculation of the stromal cells, the vessel is incubated in an appropriate nutrient medium under conditions that are metabolically favorable for the growth of the cells. As used herein, the phrase 'metabolically favorable conditions" refers to conditions that promote cell division. Such conditions include growth in nutrient medium at 37°C in a 5% CO2 incubator with greater than 90% humidity. Many commercially available media, such as RPMI 1640, Fisher's, Iscove's, McCoy's, Dulbecco's Modified Eagle's Medium, etc., and the like, which may or may not be supplemented with serum, may be suitable for use as nutrient medium. Antibiotics such as penicillin and streptomycin may also be included. In preferred embodiments, the stromal cells are cultured first, for a brief period of time (e.g., at least 1 hour), in the presence of 10% serum. The medium is then changed to a serum-free medium in order to minimize the number of extraeneous (i.e. nonstromal-cell derived) agents that would be present in the medium under continuous culture. The serum-free stromal-cell medium is therefore going to become a stromal-cell conditioned medium (supernatant) upon the continuous incubationof the stromal-cells.
  • Newly inoculated cultures may be allowed to grow under metabolically favorable conditions for between about 3-6 cell cycles. A cell cycle, as defined herein, is the length of time between mitoses. Preferably, the stromal-cells are grown to confluency (for example, at a concentration of of 107 cells/10 ml medium in suspension).
  • The supernatant from the culture of stromal-cells is then isolated. Typically, the supernatant is removed by aspiration or by centrifugation of the cell culture to remove the cells. The cultures can also be filtered to remove cells and cell-debris. The supernatant (which is suspected of containing a fugetactic agent) is then fractionated into a plurality of fractions, and a fraction from the plurality of fractions is contacted with a cell with migratory capacity. A "cell with migratory capacity" is a eukaryotic cell that includes, but is not limited to, a cell of hematopoietic origin, a cell of neural origin, a cell of epithelial origin, a cell involved in angiogenesis, a cell of mesenchymal origin, an embryonic stem cell, or a germ cell. Such cells with migratory capacity can respond to a variety of chemotactic signals, including chemoattractive and chemorepulsive (fugetactic) signals. The response typically involves changes in the actin cytoskeleton.
  • Cells of "hematopoietic origin" include, but are not limited to, pluripotent stem cells, multipotent progenitor cells and/or progenitor cells committed to specific hematopoietic lineages. The progenitor cells committed to specific hematopoietic lineages may be of T cell lineage, B cell lineage, dendritic cell lineage, Langerhans cell lineage and/or lymphoid tissue-specific macrophage cell lineage. The hematopoietic cells may be derived from a tissue such as bone marrow, peripheral blood (including mobilized peripheral blood), umbilical cord blood, placental blood, fetal liver, embryonic cells (including embryonic stem cells), aortal-gonadal-mesonephros derived cells, and lymphoid soft tissue. Lymphoid soft tissue includes the thymus, spleen, liver, lymph node, skin, tonsil and Peyer's patches. In other embodiments, the "hematopoietic origin" cells may be derived from in vitro cultures of any of the foregoing cells, and in particular in vitro cultures of progenitor cells.
  • Cells of neural origin, include neurons and glia, and/or cells of both central and peripheral nervous tissue that express RR/B (see, U.S. Patent 5,863,744, entitled: "Neural cell protein marker RR/B and DNA encoding same," issued January 26, 1999, to Avraham, et al. ).
  • Cells of epithelial origin, include cells of a tissue that covers and lines the free surfaces of the body. Such epithelial tissue includes cells of the skin and sensory organs, as well as the specialized cells lining the blood vessels, gastrointestinal tract, air passages, ducts of the kidneys and endocrine organs.
  • Cells of mesenchymal origin include cells that express typical fibroblast markers such as collagen, vimentin and fibronectin.
  • Cells involved in angiogenesis are cells that are involved in blood vessel formation and include cells of epithelial origin and cells of mesenchymal origin.
  • An embryonic stem cell is a cell that can give rise to cells of all lineages; it also has the capacity to self-renew.
  • A germ cell is a cell specialised to produce haploid gametes. It is a cell further differentiated than a stem cell, that can still give rise to more differentiated germ-line cells.
  • Movement of the cell with migratory capacity is then measured according to any of the methods decribed elsewhere herein (e.g., see Examples), and a person of ordinary skill in the art may determine whether the movement of the cell with migratory capacity is movement away from the fraction. Such movement of the cell with migratory capacity away from the fraction is indicative of the presence of a fugetactic agent in the fraction.
  • The fugetactic agent (stromal or other -cell isolate)-containing supernatant can be fractionated according to standard chromatographic procedures to facilitate isolation of the fugetactic polypeptide (thymic stromal or other -cell isolate). One of ordinary skill in the art will be familiar with such procedures that include, but are not limited to, size-exclusion chromatography, FPLC, HPLC, gel filtration chromatography, ion-exchange chromatography, hydrophobic chromatography, immune-affinity chromatography, etc. (see also the Examples section).
  • The stromal-cells may be thymic stromal-cells and the cell with migratory capacity is a hematopoietic cell. The stromal-cells may be isolated from an immune-privileged site/tissue. An "immune-privileged tissue" is a tissue with immune-cell tolerance and includes, but is not limited to, brain tissue, central nervous system (CNS), testes, placenta, or ocular tissue (e.g., aqueous humour).
  • Other fugetactic molecules can be isolated from different tissues or biological fluid homogenates or supernatants, or other biological materials, using the foregoing methodology. In preferred embodiments, the biological fluid includes, but is not limited to, synovial fluid, cerebral spinal fluid, fallopian tube fluid, seminal fluid, ocular fluid, pericardial fluid, pleural fluid, inflammatory exudate and ascitic fluid.
  • The agent suspected of being a fugetactic agent may be an agent present in a tumor cell culture supernatant, tumor cell eluate, and/or tumor cell lysate (see also the Examples section). The tumor cell may be of a cancer or tumor type thought to escape immune recognition. Such cancers or tumors may be of the folowing origin: biliary tract cancer; brain cancer, including glioblastomas and medulloblastomas; breast cancer; cervical cancer; choriocarcinoma; colon cancer; endometrial cancer; esophageal cancer; gastric cancer; hematological neoplasms, including acute lymphocytic and myelogenous leukemia; multiple myeloma; AIDS associated leukemias and adult T-cell leukemia lymphoma; intraepithelial neoplasms, including Bowen's disease and Paget's disease; liver cancer (hepatocarcinoma); lung cancer; lymphomas, including Hodgkin's disease and lymphocytic lymphomas; neuroblastomas; oral cancer, including squamous cell carcinoma; ovarian cancer, including those arising from epithelial cells, stromal cells, germ cells and mesenchymal cells; pancreas cancer; prostate cancer; rectal cancer; sarcomas, including leiomyosarcoma, rhabdomyosarcoma, liposarcoma, fibrosarcoma and osteosarcoma; skin cancer, including melanoma, Kaposi's sarcoma, basocellular cancer and squamous cell cancer; testicular cancer, including germinal tumors (seminoma, non-seminoma[teratomas, choriocarcinomas]), stromal tumors and germ cell tumors; thyroid cancer, including thyroid adenocarcinoma and medullar carcinoma; and renal cancer including adenocarcinoma and Wilms tumor. In important embodiments, cancers or tumors escaping immune recognition include glioma, colon carcinoma, colorectal cancer, lymphoid cell-derived leukemia, choriocarcinoma, and melanoma.
  • In any of the foregoing description, the cell with migratory capacity is a hematopoietic cell. The hematopoietic cell may be an immune cell.
  • There is also described herein a method of repelling immune cells from a material surface. "Material surfaces" as used herein, include, but are not limited to, dental and orthopedic prosthetic implants, artificial valves, and organic implantable tissue such as a stent, allogeneic and/or xenogeneic tissue, organ and/or vasculature.
  • Implantable prosthetic devices have been used in the surgical repair or replacement of internal tissue for many years. Orthopedic implants include a wide variety of devices, each suited to fulfill particular medical needs. Examples of such devices are hip joint replacement devices, knee joint replacement devices, shoulder joint replacement devices, and pins, braces and plates used to set fractured bones. Some contemporary orthopedic and dental implants, use high performance metals such as cobalt-chrome and titanium alloy to achieve high strength. These materials are readily fabricated into the complex shapes typical of these devices using mature metal working techniques including casting and machining.
  • The material surface is coated with an amount of a fugetactic agent effective to repel immune cells. In important embodiments, the material surface is part of an implant. In important embodiments, in addition to a fugetactic agent, the material surface may also be coated with a cell-growth potentiating agent, an anti-infective agent, and/or an antiinflammatory agent.
  • A cell-growth potentiating agent as used herein is an agent which stimulates growth of a cell and includes growth factors such as PDGF, EGF, FGF, TGF, NGF, CNTF, and GDNF.
  • An anti-infectious agent as used herein is an agent which reduces the activity of or kills a microorganism and includes: Aztreonam; Chlorhexidine Gluconate; Imidurea; Lycetamine; Nibroxane; Pirazmonam Sodium; Propionic Acid; Pyrithione Sodium; Sanguinarium Chloride; Tigemonam Dicholine; Acedapsone; Acetosulfone Sodium; Alamecin; Alexidine; Amdinocillin; Amdinocillin Pivoxil; Amicycline; Amifloxacin; Amifloxacin Mesylate; Amikacin; Amikacin Sulfate; Aminosalicylic acid; Aminosalicylate sodium; Amoxicillin; Amphomycin; Ampicillin; Ampicillin Sodium; Apalcillin Sodium; Apramycin; Aspartocin; Astromicin Sulfate; Avilamycin; Avoparcin; Azithromycin; Azlocillin; Azlocillin Sodium; Bacampicillin Hydrochloride; Bacitracin; Bacitracin Methylene Disalicylate; Bacitracin Zinc; Bambermycins; Benzoylpas Calcium; Berythromycin; Betamicin Sulfate; Biapenem; Biniramycin; Biphenamine Hydrochloride; Bispyrithione Magsulfex; Butikacin; Butirosin Sulfate; Capreomycin Sulfate; Carbadox; Carbenicillin Disodium; Carbenicillin Indanyl Sodium; Carbenicillin Phenyl Sodium; Carbenicillin Potassium; Carumonam Sodium; Cefaclor; Cefadroxil; Cefamandole; Cefamandole Nafate; Cefamandole Sodium; Cefaparole; Cefatrizine; Cefazaflur Sodium; Cefazolin; Cefazolin Sodium; Cefbuperazone; Cefdinir; Cefepime; Cefepime Hydrochloride; Cefetecol; Cefixime; Cefmenoxime Hydrochloride; Cefmetazole; Cefmetazole Sodium; Cefonicid Monosodium; Cefonicid Sodium; Cefoperazone Sodium; Ceforanide; Cefotaxime Sodium; Cefotetan; Cefotetan Disodium; Cefotiam Hydrochloride; Cefoxitin; Cefoxitin Sodium; Cefpimizole; Cefpimizole Sodium; Cefpiramide; Cefpiramide Sodium; Cefpirome Sulfate; Cefpodoxime Proxetil; Cefprozil; Cefroxadine; Cefsulodin Sodium; Ceftazidime; Ceftibuten; Ceftizoxime Sodium; Ceftriaxone Sodium; Cefuroxime; Cefuroxime Axetil; Cefuroxime Pivoxetil; Cefuroxime Sodium; Cephacetrile Sodium; Cephalexin; Cephalexin Hydrochloride; Cephaloglycin; Cephaloridine; Cephalothin Sodium; Cephapirin Sodium; Cephradine; Cetocycline Hydrochloride; Cetophenicol; Chloramphenicol; Chloramphenicol Palmitate; Chloramphenicol Pantothenate Complex; Chloramphenicol Sodium Succinate; Chlorhexidine Phosphanilate; Chloroxylenol; Chlortetracycline Bisulfate; Chlortetracycline Hydrochloride; Cinoxacin; Ciprofloxacin; Ciprofloxacin Hydrochloride; Cirolemycin; Clarithromycin; Clinafloxacin Hydrochloride; Clindamycin; Clindamycin Hydrochloride; Clindamycin Palmitate Hydrochloride; Clindamycin Phosphate; Clofazimine; Cloxacillin Benzathine; Cloxacillin Sodium; Cloxyquin; Colistimethate Sodium; Colistin Sulfate; Coumermycin; Coumermycin Sodium; Cyclacillin; Cycloserine; Dalfopristin; Dapsone; Daptomycin; Demeclocycline; Demeclocycline Hydrochloride; Demecycline; Denofungin; Diaveridine; Dicloxacillin; Dicloxacillin Sodium; Dihydrostreptomycin Sulfate; Dipyrithione; Dirithromycin; Doxycycline; Doxycycline Calcium; Doxycycline Fosfatex; Doxycycline Hyclate; Droxacin Sodium; Enoxacin; Epicillin; Epitetracycline Hydrochloride; Erythromycin; Erythromycin Acistrate; Erythromycin Estolate; Erythromycin Ethylsuccinate; Erythromycin Gluceptate; Erythromycin Lactobionate; Erythromycin Propionate; Erythromycin Stearate; Ethambutol Hydrochloride; Ethionamide; Fleroxacin; Floxacillin; Fludalanine; Flumequine; Fosfomycin; Fosfomycin Tromethamine; Fumoxicillin; Furazolium Chloride; Furazolium Tartrate; Fusidate Sodium; Fusidic Acid; Gentamicin Sulfate; Gloximonam; Gramicidin; Haloprogin; Hetacillin; Hetacillin Potassium; Hexedine; Ibafloxacin; Imipenem; Isoconazole; Isepamicin; Isoniazid; Josamycin; Kanamycin Sulfate; Kitasamycin; Levofuraltadone; Levopropylcillin Potassium; Lexithromycin; Lincomycin; Lincomycin Hydrochloride; Lomefloxacin; Lomefloxacin Hydrochloride; Lomefloxacin Mesylate; Loracarbef; Mafenide; Meclocycline; Meclocycline Sulfosalicylate; Megalomicin Potassium Phosphate; Mequidox; Meropenem; Methacycline; Methacycline Hydrochloride; Methenamine; Methenamine Hippurate; Methenamine Mandelate; Methicillin Sodium; Metioprim; Metronidazole Hydrochloride; Metronidazole Phosphate; Mezlocillin; Mezlocillin Sodium; Minocycline; Minocycline Hydrochloride; Mirincamycin Hydrochloride; Monensin; Monensin Sodium; Nafcillin Sodium; Nalidixate Sodium; Nalidixic Acid; Natamycin; Nebramycin; Neomycin Palmitate; Neomycin Sulfate; Neomycin Undecylenate; Netilmicin Sulfate; Neutramycin; Nifuradene; Nifuraldezone; Nifuratel; Nifuratrone; Nifurdazil; Nifurimide; Nifurpirinol; Nifurquinazol; Nifurthiazole; Nitrocycline; Nitrofurantoin; Nitromide; Norfloxacin; Novobiocin Sodium; Ofloxacin; Ormetoprim; Oxacillin Sodium; Oximonam; Oximonam Sodium; Oxolinic Acid; Oxytetracycline; Oxytetracycline Calcium; Oxytetracycline Hydrochloride; Paldimycin; Parachlorophenol; Paulomycin; Pefloxacin; Pefloxacin Mesylate; Penamecillin; Penicillin G Benzathine; Penicillin G Potassium; Penicillin G Procaine; Penicillin G Sodium; Penicillin V; Penicillin V Benzathine; Penicillin V Hydrabamine; Penicillin V Potassium; Pentizidone Sodium; Phenyl Aminosalicylate; Piperacillin Sodium; Pirbenicillin Sodium; Piridicillin Sodium; Pirlimycin Hydrochloride; Pivampicillin Hydrochloride; Pivampicillin Pamoate; Pivampicillin Probenate; Polymyxin B Sulfate; Porfiromycin; Propikacin; Pyrazinamide; Pyrithione Zinc; Quindecamine Acetate; Quinupristin; Racephenicol; Ramoplanin; Ranimycin; Relomycin; Repromicin; Rifabutin; Rifametane: Rifamexil; Rifamide; Rifampin; Rifapentine; Rifaximin; Rolitetracycline; Rolitetracycline Nitrate; Rosaramicin; Rosaramicin Butyrate; Rosaramicin Propionate; Rosaramicin Sodium Phosphate; Rosaramicin Stearate; Rosoxacin; Roxarsone; Roxithromycin; Sancycline; Sanfetrinem Sodium; Sarmoxicillin; Sarpicillin; Scopafungin; Sisomicin; Sisomicin Sulfate; Sparfloxacin; Spectinomycin Hydrochloride; Spiramycin; Stallimycin Hydrochloride; Steffimycin; Streptomycin Sulfate; Streptonicozid; Sulfabenz; Sulfabenzamide; Sulfacetamide; Sulfacetamide Sodium; Sulfacytine; Sulfadiazine; Sulfadiazine Sodium; Sulfadoxine; Sulfalene; Sulfamerazine; Sulfameter; Sulfamethazine; Sulfamethizole; Sulfamethoxazole; Sulfamonomethoxine; Sulfamoxole; Sulfanilate Zinc; Sulfanitran; Sulfasalazine; Sulfasomizole; Sulfathiazole; Sulfazamet; Sulfisoxazole; Sulfisoxazole Acetyl; Sulfisoxazole Diolamine; Sulfomyxin; Sulopenem; Sultamicillin; Suncillin Sodium; Talampicillin Hydrochloride; Teicoplanin; Temafloxacin Hydrochloride; Temocillin; Tetracycline; Tetracycline Hydrochloride; Tetracycline Phosphate Complex; Tetroxoprim; Thiamphenicol; Thiphencillin Potassium; Ticarcillin Cresyl Sodium; Ticarcillin Disodium; Ticarcillin Monosodium; Ticlatone; Tiodonium Chloride; Tobramycin; Tobramycin Sulfate; Tosufloxacin; Trimethoprim; Trimethoprim Sulfate; Trisulfapyrimidines; Troleandomycin; Trospectomycin Sulfate; Tyrothricin; Vancomycin; Vancomycin Hydrochloride; Virginiamycin; Zorbamycin; Difloxacin Hydrochloride; Lauryl Isoquinolinium Bromide; Moxalactam Disodium; Omidazole; Pentisomicin; and Sarafloxacin Hydrochloride.
  • Antiinflammatory agents are as described below.
  • There is also described herein a method of inhibiting migration of immune cells to a specific site in a subject is provided. The method involves locally administering to a specific site in a subject in need of such treatment a fugetactic agent in an amount effective to inhibit migration of immune cells to the specific site in a subject.
  • There is also described herein a method of inhibiting migration of immune cells to a site of inflammation in the subject. "Inflammation" as used herein, is a localised protective response elicited by a foreign (non-self) antigen, and/or by an injury or destruction of tissue(s), which serves to destroy, dilute or sequester the foreign antigen, the injurious agent, and/or the injured tissue. Inflammation occurs when tissues are injured by viruses, bacteria, trauma, chemicals, heat, cold, or any other harmful stimuli. In such instances, the classic weapons of the immune system (T cells, B cells, macrophages) interface with cells and soluble products that are mediators of inflammatory responses (neutrophils, eosinophils, basophils, kinin and coagulation systems, and complement cascade).
  • A typical inflammatory response is characterized by (i) migration of leukocytes at the site of antigen (injury) localization; (ii) specific and nonspecific recognition of "foreign" and other (necrotic/injured tissue) antigens mediated by B and T lymphocytes, macrophages and the alternative complement pathway; (iii) amplification of the inflammatory response with the recruitment of specific and nonspecific effector cells by complement components, lymphokines and monokines, kinins, arachidonic acid metabolites, and mast cell/basophil products; and (iv) macrophage, neutrophil and lymphocyte participation in antigen destruction with ultimate removal of antigen particles (injured tissue) by phagocytosis. The ability of the immune system to discriminate between "self' and "non-self'(foreign) antigens is therefore vital to the functioning of the immune system as a specific defense against "non-self" antigens.
  • "Non-self" antigens are those antigens on substances entering a subject, or exist in a subject but are detectably different or foreign from the subject's own constituents, whereas "self" antigens are those which, in the healthy subject, are not detectably different or foreign from its own constituents. However, under certain conditions, including in certain disease states, an individual's immune system will identify its own constituents as "non-self," and initiate an immune response against "self-antigens," at times causing more damage or discomfort as from, for example, an invading microbe or foreign material, and often producing serious illness in a subject.
  • The inflammation may be caused by an immune response against "self-antigen," and the subject in need of treatment according to the invention has an autoimmune disease. "Autoimmune disease" as used herein, results when a subject's immune system attacks its own organs or tissues, producing a clinical condition associated with the destruction of that tissue, as exemplified by diseases such as rheumatoid arthritis, uveitis, insulin-dependent diabetes mellitus, hemolytic anemias, rheumatic fever, Crohn's disease, Guillain-Barre syndrome, psoriasis, thyroiditis, Graves' disease, myasthenia gravis, glomerulonephritis, autoimmune hepatitis, multiple sclerosis, systemic lupus erythematosus, etc.
  • Autoimmune disease may be caused by a genetic predisposition alone, by certain exogenous agents (e.g., viruses, bacteria, chemical agents, etc.), or both. Some forms of autoimmunity arise as the result of trauma to an area usually not exposed to lymphocytes, such as neural tissue or the lens of the eye. When the tissues in these areas become exposed to lymphocytes, their surface proteins can act as antigens and trigger the production of antibodies and cellular immune responses which then begin to destroy those tissues. Other autoimmune diseases develop after exposure of a subject to antigens which are antigenically similar to, that is cross-reactive with, the subject's own tissue. In rheumatic fever, for example, an antigen of the streptococcal bacterium, which causes rheumatic fever, is cross-reactive with parts of the human heart. The antibodies cannot differentiate between the bacterial antigens and the heart muscle antigens, consequently cells with either of those antigens can be destroyed.
  • Other autoimmune diseases, for example, insulin-dependent diabetes mellitus (involving the destruction of the insulin producing beta-cells of the islets of Langerhans), multiple sclerosis (involving the destruction of the conducting fibers of the nervous system) and rheumatoid arthritis (involving the destruction of the joint-lining tissue), are characterized as being the result of a mostly cell-mediated autoimmune response and appear to be due primarily to the action of T cells (See, Sinha et al., Science, 1990, 248:1380). Yet others, such as myesthenia gravis and systemic lupus erythematosus, are characterized as being the result of primarily a humoral autoimmune response. Nevertheless, inhibition of migration of immune cells to a specific site of inflammation involved in any of the foregoing conditions according to the invention, is beneficial to the subject since it inhibits escalation of the inflammatory response, protecting the specific site (e.g., tissue) involved, from "self-damage." In preffered embodiments, the subject has rheumatoid arthritis, multiple sclerosis, or uveitis.
  • The inflammation may be caused by an immune response against "non-self-antigens" (including antigens of necrotic self-material), and the subject in need of treatment according to the invention is a transplant recipient, has atherosclerosis, has suffered a myocardial infarction and/or an ischemic stroke, has an abscess, and/or has myocarditis. This is because after cell (or organ) transplantation, or after myocardial infarction or ischemic stroke, certain antigens from the transplanted cells (organs), or necrotic cells from the heart or the brain, can stimulate the production of immune lymphocytes and/or autoantibodies, which later participate in inflammation/rejection (in the case of a transplant), or attack cardiac or brain target cells causing inflammation and aggravating the condition (Johnson et al., Sem. Nuc. Med. 1989, 19:238; Leinonen et al., Microbiol. Path.,1990, 9:67; Montalban et al., Stroke, 1991, 22:750).
  • There is also described herein a method of enhancing an immune response in a subject having a condition that involves a specific site, is provided. The method involves locally administering to a specific site in a subject in need of such treatment an anti-fugetactic agent in an amount effective to inhibit immune cell-specific fugetactic activity at a specific site in the subject. The specific site may be a site of a pathogenic infection. Efficient recruitment of immune cells to help eliminate the infection is therefore beneficial.
  • The specific site may be a germ cell containing site. In this case the recruitment of immune cells to these specific sites will help eliminate unwanted germ cells, and/or implanted and nonimplanted embryos. Co-administration of contraceptive agents other than anti-fugetactic agents is also described herein. Non-anti-fugetactic contraceptive agents are well known in the art.
  • In further embodiments, the specific site is an area immediately surrounding a tumor. Since most of the known tumors escape immune recognition, it is beneficial to enhance the migration of immune cells to the tumor site. In further embodiments, co-administration of anti-cancer agents other than anti-fugetactic agents is also provided. Non-anti-fugetactic anti-cancer agents include: Acivicin; Aclarubicin; Acodazole Hydrochloride; Acronine; Adozelesin; Aldesleukin; Altretamine; Ambomycin; Ametantrone Acetate; Aminoglutethimide; Amsacrine; Anastrozole; Anthramycin; Asparaginase; Asperlin; Azacitidine; Azetepa; Azotomycin; Batimastat; Benzodepa; Bicalutamide; Bisantrene Hydrochloride; Bisnafide Dimesylate; Bizelesin; Bleomycin Sulfate; Brequinar Sodium; Bropirimine; Busulfan; Cactinomycin; Calusterone; Caracemide; Carbetimer; Carboplatin; Carmustine; Carubicin Hydrochloride; Carzelesin; Cedefingol; Chlorambucil; Cirolemycin; Cisplatin; Cladribine; Crisnatol Mesylate; Cyclophosphamide; Cytarabine; Dacarbazine; Dactinomycin; Daunorubicin Hydrochloride; Decitabine; Dexormaplatin; Dezaguanine; Dezaguanine Mesylate; Diaziquone; Docetaxel; Doxorubicin; Doxorubicin Hydrochloride; Droloxifene; Droloxifene Citrate; Dromostanolone Propionate; Duazomycin; Edatrexate; Eflornithine Hydrochloride; Elsamitrucin; Enloplatin; Enpromate; Epipropidine; Epirubicin Hydrochloride; Erbulozole; Esorubicin Hydrochloride; Estramustine; Estramustine Phosphate Sodium; Etanidazole; Etoposide; Etoposide Phosphate; Etoprine; Fadrozole Hydrochloride; Fazarabine; Fenretinide; Floxuridine; Fludarabine Phosphate; Fluorouracil; Flurocitabine; Fosquidone; Fostriecin Sodium; Gemcitabine; Gemcitabine Hydrochloride; Hydroxyurea; Idarubicin Hydrochloride; Ifosfamide; Ilmofosine; Interferon Alfa-2a; Interferon Alfa-2b; Interferon Alfa-n1; Interferon Alfa-n3; Interferon Beta-I a; Interferon Gamma-I b; Iproplatin; Irinotecan Hydrochloride; Lanreotide Acetate; Letrozole; Leuprolide Acetate; Liarozole Hydrochloride; Lometrexol Sodium; Lomustine; Losoxantrone Hydrochloride; Masoprocol; Maytansine; Mechlorethamine Hydrochloride; Megestrol Acetate; Melengestrol Acetate; Melphalan; Menogaril; Mercaptopurine; Methotrexate; Methotrexate Sodium; Metoprine; Meturedepa; Mitindomide; Mitocarcin; Mitocromin; Mitogillin; Mitomalcin; Mitomycin; Mitosper; Mitotane; Mitoxantrone Hydrochloride; Mycophenolic Acid; Nocodazole; Nogalamycin; Ormaplatin; Oxisuran; Paclitaxel; Pegaspargase; Peliomycin; Pentamustine; Peplomycin Sulfate; Perfosfamide; Pipobroman; Piposulfan; Piroxantrone Hydrochloride; Plicamycin; Plomestane; Podofilox; Porfimer Sodium; Porfiromycin; Prednimustine; Procarbazine Hydrochloride; Puromycin; Puromycin Hydrochloride; Pyrazofurin; Riboprine; Rogletimide; Safingol; Safingol Hydrochloride; Semustine; Simtrazene; Sparfosate Sodium; Sparsomycin; Spirogermanium Hydrochloride; Spiromustine; Spiroplatin; Streptonigrin; Streptozocin; Sulofenur; Talisomycin; Taxotere; Tecogalan Sodium; Tegafur; Teloxantrone Hydrochloride; Temoporfin; Teniposide; Teroxirone; Testolactone; Thiamiprine; Thioguanine; Thiotepa; Tiazofurin; Tirapazamine; Topotecan Hydrochloride; Toremifene Citrate; Trestolone Acetate; Triciribine Phosphate; Trimetrexate; Trimetrexate Glucuronate; Triptorelin; Tubulozole Hydrochloride; Uracil Mustard; Uredepa; Vapreotide; Verteporfin; Vinblastine Sulfate; Vincristine Sulfate; Vindesine; Vindesine Sulfate; Vinepidine Sulfate; Vinglycinate Sulfate; Vinleurosine Sulfate; Vinorelbine Tartrate; Vinrosidine Sulfate; Vinzolidine Sulfate; Vorozole; Zeniplatin; Zinostatin; Zorubicin Hydrochloride.
  • There is also described herein a method of inhibiting tumor cell metastasis in a subject. The method involves locally administering to a tumor site in a subject in need of such treatment an anti-fugetactic agent in an amount effective to inhibit metastasis of tumor cells from the tumor site in the subject.
  • Co-administration of anti-cancer agents other than anti-fugetactic agents is also described. Anti-cancer agents are as described above.
  • There is also described herein a method of inhibiting endothelial cell migration to a tumor site in a subject. The method involves locally administering to an area surrounding a tumor site in a subject in need of such treatment a fugetactic agent in an amount effective to inhibit endothelial cell migration to the tumor site in the subject.
  • The area surrounding the tumor site may not be immediate to the tumor site. Important fugetactic agents are as described above.
  • There is also described herein a method of contraception in a subject, The method involves administering to a subject in need of such treatment, an anti-fugetactic agent in an amount effective to inhibit migration of germ cells in the subject.
  • The administration may be local to a germ cell-containing site of the subject.
  • There is also described a method of treating infertility and premature labor, including premature delivery and impending miscarriage. The method involves administering to a subject in need of such treatment a fugetactic agent in an amount effective to inhibit immune cells from migrating close to a germ cell (including an egg, a sperm, a fertilized egg, or an implanted embryo) in the subject. The administration may be local to a germ cell-containing site of the subject.
  • The foregoing methods of therapy may include co-administration of a non-fugetactic agent together with a fugetactic agent of the invention that can act cooperatively, additively, or synergistically with the fugetactic agent of the invention to inhibit migration of immune cells to the site of inflammation in the subject. A fugetactic agent may be administered substantially simultaneously with a non-fugetactic agent to inhibit migration of immune cells to a site of inflammation. By "substantially simultaneously," it is meant that the fugetactic agent is locally administered to the subject close enough in time with the administration of the non-fugetactic agent, whereby the non-fugetactic agent may exert a potentiating effect on migration inhibiting activity of the fugetactic agent. Thus, by substantially simultaneously it is meant that the fugetactic agent is administered before, at the same time, and/or after the administration of the non-fugetactic agent. The fugetactic agent can be administered as a polypeptide, and/or a nucleic acid which expresses a fugetactic agent.
  • The non-fugetactic agents may be immunosuppressants. Such immunosuppressants include: Azathioprine; Azathioprine Sodium; Cyclosporine; Daltroban; Gusperimus Trihydrochloride; Sirolimus; Tacrolimus.
  • The non-fugetactic agents may be anti-inflammatory agents. Such anti-inflammatory agents include: Alclofenac; Alclometasone Dipropionate; Algestone Acetonide; Alpha Amylase; Amcinafal; Amcinafide; Amfenac Sodium; Amiprilose Hydrochloride; Anakinra; Anirolac; Anitrazafen; Apazone; Balsalazide Disodium; Bendazac; Benoxaprofen; Benzydamine Hydrochloride; Bromelains; Broperamole; Budesonide; Carprofen; Cicloprofen; Cintazone; Cliprofen; Clobetasol Propionate; Clobetasone Butyrate; Clopirac; Cloticasone Propionate; Cormethasone Acetate; Cortodoxone; Deflazacort; Desonide; Desoximetasone; Dexamethasone Dipropionate; Diclofenac Potassium; Diclofenac Sodium; Diflorasone Diacetate; Diflumidone Sodium; Diflunisal; Difluprednate; Diftalone; Dimethyl Sulfoxide; Drocinonide; Endrysone; Enlimomab; Enolicam Sodium; Epirizole; Etodolac; Etofenamate; Felbinac; Fenamole; Fenbufen; Fenclofenac; Fenclorac; Fendosal; Fenpipalone; Fentiazac; Flazalone; Fluazacort; Flufenamic Acid; Flumizole; Flunisolide Acetate; Flunixin; Flunixin Meglumine; Fluocortin Butyl; Fluorometholone Acetate; Fluquazone; Flurbiprofen; Fluretofen; Fluticasone Propionate; Furaprofen; Furobufen; Halcinonide; Halobetasol Propionate; Halopredone Acetate; Ibufenac; Ibuprofen; Ibuprofen Aluminum; Ibuprofen Piconol; Ilonidap; Indomethacin; Indomethacin Sodium; Indoprofen; Indoxole; Intrazole; Isoflupredone Acetate; Isoxepac; Isoxicam; Ketoprofen; Lofemizole Hydrochloride; Lomoxicam; Loteprednol Etabonate; Meclofenamate Sodium; Meclofenamic Acid; Meclorisone Dibutyrate; Mefenamic Acid; Mesalamine; Meseclazone; Methylprednisolone Suleptanate; Morniflumate; Nabumetone; Naproxen; Naproxen Sodium; Naproxol; Nimazone; Olsalazine Sodium; Orgotein; Orpanoxin; Oxaprozin; Oxyphenbutazone; Paranyline Hydrochloride; Pentosan Polysulfate Sodium; Phenbutazone Sodium Glycerate; Pirfenidone; Piroxicam; Piroxicam Cinnamate; Piroxicam Olamine; Pirprofen; Prednazate; Prifelone; Prodolic Acid; Proquazone; Proxazole; Proxazole Citrate; Rimexolone; Romazarit; Salcolex; Salnacedin; Salsalate; Sanguinarium Chloride; Seclazone; Sermetacin; Sudoxicam; Sulindac; Suprofen; Talmetacin; Talniflumate; Talosalate; Tebufelone; Tenidap; Tenidap Sodium; Tenoxicam; Tesicam; Tesimide; Tetrydamine; Tiopinac; Tixocortol Pivalate; Tolmetin; Tolmetin Sodium; Triclonide; Triflumidate; Zidometacin; Zomepirac Sodium.
  • The compositions, as described above, are administered in effective amounts. The effective amount will depend upon the mode of administration, the particular condition being treated and the desired outcome. It will also depend upon, as discussed above, the stage of the condition, the age and physical condition of the subject, the nature of concurrent therapy, if any, and like factors well known to the medical practitioner. For therapeutic applications, it is that amount sufficient to achieve a medically desirable result. In some cases this is a local (site-specific) reduction of inflammation. In other cases, it is inhibition of tumor growth and/or metastasis.
  • Generally, doses of active compounds of the present invention would be from about 0.01 mg/kg per day to 1000 mg/kg per day. It is expected that doses ranging from 50-500 mg/kg will be suitable. A variety of administration routes are available. The methods of the invention, generally speaking, may be practiced using any mode of administration that is medically acceptable, meaning any mode that produces effective levels of the active compounds without causing clinically unacceptable adverse effects. Such modes of administration include oral, rectal, topical, nasal, interdermal, or parenteral routes. The term "parenteral" includes subcutaneous, intravenous, intramuscular, or infusion. Intravenous or intramuscular routes are not particularly suitable for long-term therapy and prophylaxis. They could, however, be preferred in emergency situations. Oral administration will be preferred for prophylactic treatment because of the convenience to the patient as well as the dosing schedule. When peptides are used therapeutically, in certain embodiments a desirable route of administration is by pulmonary aerosol. Techniques for preparing aerosol delivery systems containing peptides are well known to those of skill in the art. Generally, such systems should utilize components which will not significantly impair the biological properties of the antibodies, such as the paratope binding capacity (see, for example, Sciarra and Cutie, "Aerosols," in Remington's Pharmaceutical Sciences, 18th edition, 1990, pp 1694-1712 ). Those of skill in the art can readily determine the various parameters and conditions for producing antibody or peptide aerosols without resort to undue experimentation.
  • Compositions suitable for oral administration may be presented as discrete units, such as capsules, tablets, lozenges, each containing a predetermined amount of the active agent. Other compositions include suspensions in aqueous liquids or non-aqueous liquids such as a syrup, elixir or an emulsion.
  • Preparations for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions, and emulsions. Examples of non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate. Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media. Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's or fixed oils. Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers (such as those based on Ringer's dextrose), and the like. Preservatives and other additives may also be present such as, for example, antimicrobials, anti-oxidants, chelating agents, and inert gases and the like. Lower doses will result from other forms of administration, such as intravenous administration. In the event that a response in a subject is insufficient at the initial doses applied, higher doses (or effectively higher doses by a different, more localized delivery route) may be employed to the extent that patient tolerance permits. Multiple doses per day are contemplated to achieve appropriate systemic levels of compounds.
  • The fugetactic agents, fugetactic binding agents, or fragments thereof may be combined, optionally, with a pharmaceutically-acceptable carrier. The term "pharmaceutically-acceptable carrier" as used herein means one or more compatible solid or liquid filler, diluents or encapsulating substances which are suitable for administration into a human. The term "carrier" denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application. The components of the pharmaceutical compositions also are capable of being co-mingled with the molecules of the present invention, and with each other, in a manner such that there is no interaction which would substantially impair the desired pharmaceutical efficacy.
  • There is also described herein pharmaceutical compositions of fugetactic agents and anti-fugetactic agents.
  • When administered, the pharmaceutical preparations described herein are applied in pharmaceutically-acceptable amounts and in pharmaceutically-acceptably compositions. Such preparations may routinely contain salt, buffering agents, preservatives, compatible carriers, and optionally other therapeutic agents. When used in medicine, the salts should be pharmaceutically acceptable, but non-pharmaceutically acceptable salts may conveniently be used to prepare pharmaceutically-acceptable salts thereof and are not excluded from the scope of the invention. Such pharmacologically and pharmaceutically-acceptable salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulfuric, nitric, phosphoric, maleic, acetic, salicylic, citric, formic, malonic, succinic, and the like. Also, pharmaceutically-acceptable salts can be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium or calcium salts.
  • Fugetactic molecules (nucleic acids or polypeptides) preferably are produced recombinantly, although such molecules may be isolated from biological extracts. Recombinantly produced fugetactic agents such as SDF-1α polypeptides, include chimeric proteins comprising a fusion of a SDF-1α protein with another polypeptide, e.g., a polypeptide capable of providing or enhancing protein-protein binding, sequence specific nucleic acid binding (such as GAL4), enhancing stability of the SDF-1α polypeptide under assay conditions, or providing a detectable moiety, such as green fluorescent protein. A polypeptide fused to a SDF-1α polypeptide or fragment may also provide means of readily detecting the fusion protein, e.g., by immunological recognition or by fluorescent labeling.
  • Various techniques may be employed for introducing nucleic acids of the invention (SDF-1α sense and anti-sense, dominant negative) into cells, depending on whether the nucleic acids are introduced in vitro or in vivo in a host. Such techniques include transfection of nucleic acid-CaPO4 precipitates, transfection of nucleic acids associated with DEAE, transfection with a retrovirus including the nucleic acid of interest, liposome mediated transfection, and the like. For certain uses, it is preferred to target the nucleic acid to particular cells. In such instances, a vehicle used for delivering a nucleic acid of the invention into a cell (e.g., a retrovirus, or other virus; a liposome) can have a targeting molecule attached thereto. For example, a molecule such as an antibody specific for a surface membrane protein on the target cell or a ligand for a receptor on the target cell can be bound to or incorporated within the nucleic acid delivery vehicle. For example, where liposomes are employed to deliver the nucleic acids of the invention, proteins which bind to a surface membrane protein associated with endocytosis may be incorporated into the liposome formulation for targeting and/or to facilitate uptake. Such proteins include capsid proteins or fragments thereof tropic for a particular cell type, antibodies for proteins which undergo internalization in cycling, proteins that target intracellular localization and enhance intracellular half life, and the like. Polymeric delivery systems also have been used successfully to deliver nucleic acids into cells, as is known by those skilled in the art. Such systems even permit oral delivery of nucleic acids.
  • Other delivery systems can include time-release, delayed release or sustained release delivery systems. Such systems can avoid repeated administrations of the fugetactic agent, increasing convenience to the subject and the physician. Many types of release delivery systems are available and known to those of ordinary skill in the art. They include polymer base systems such as poly(lactide-glycolide), copolyoxalates, polycaprolactones, polyesteramides, polyorthoesters, polyhydroxybutyric acid, and polyanhydrides. Microcapsules of the foregoing polymers containing drugs are described in, for example, U.S. Patent 5,075,109 . Delivery systems also include non-polymer systems that are: lipids including sterols such as cholesterol, cholesterol esters and fatty acids or neutral fats such as mono- di- and tri-glycerides; hydrogel release systems; sylastic systems; peptide based systems; wax coatings; compressed tablets using conventional binders and excipients; partially fused implants; and the like. Specific examples include, but are not limited to: (a) erosional systems in which the anti-inflammatory agent is contained in a form within a matrix such as those described in U.S. Patent Nos. 4,452,775 , 4,667,014 , 4,748,034 and 5,239,660 and (b) difusional systems in which an active component permeates at a controlled rate from a polymer such as described in U.S. Patent Nos. 3,832,253 , and 3,854,480 .
  • A preferred delivery system is a colloidal dispersion system. Colloidal dispersion systems include lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes. A preferred colloidal system is a liposome. Liposomes are artificial membrane vessels which are useful as a delivery vector in vivo or in vitro. It has been shown that large unilamellar vessels (LUV), which range in size from 0.2 - 4.0 µm can encapsulate large macromolecules. RNA, DNA, and intact virions can be encapsulated within the aqueous interior and be delivered to cells in a biologically active form (Fraley, et al., Trends Biochem. Sci., (1981) 6:77). In order for a liposome to be an efficient gene transfer vector, one or more of the following characteristics should be present: (1) encapsulation of the gene of interest at high efficiency with retention of biological activity; (2) preferential and substantial binding to a target cell in comparison to non-target cells; (3) delivery of the aqueous contents of the vesicle to the target cell cytoplasm at high efficiency; and (4) accurate and effective expression of genetic information.
  • Liposomes may be targeted to a particular tissue by coupling the liposome to a specific ligand such as a monoclonal antibody, sugar, glycolipid, or protein. Liposomes are commercially available from Gibco BRL, for example, as LIPOFECTIN™ and LIPOFECTACE™, which are formed of cationic lipids such as N-[1-(2, 3 dioleyloxy)-propyl]-N, N, N-trimethylammonium chloride (DOTMA) and dimethyl dioctadecylammonium bromide (DDAB). Methods for making liposomes are well known in the art and have been described in many publications. Liposomes also have been reviewed by Gregoriadis, G. in Trends in Biotechnology, (1985) 3:235-241.
  • The preferred vehicle may be a biocompatible microparticle or implant that is suitable for implantation into the mammalian recipient. Exemplary bioerodible implants that are useful in accordance with this method are described in PCT International application no. PCT/US/03307 (Publication No. WO 95/24929 , entitled "Polymeric Gene Delivery System"). PCT/US/0307 describes a biocompatible, preferably biodegradable polymeric matrix for containing an exogenous gene under the control of an appropriate promoter. The polymeric matrix is used to achieve sustained release of the exogenous gene in the patient. In accordance with the instant invention, the fugetactic agents described herein are encapsulated or dispersed within the biocompatible, preferably biodegradable polymeric matrix disclosed in PCT/US/03307 .
  • The polymeric matrix preferably is in the form of a microparticle such as a microsphere (wherein a fugetactic agent is dispersed throughout a solid polymeric matrix) or a microcapsule (wherein a fugetactic agent is stored in the core of a polymeric shell). Other forms of the polymeric matrix for containing a fugetactic agent include films, coatings, gels, implants, and stents. The size and composition of the polymeric matrix device is selected to result in favorable release kinetics in the tissue into which the matrix is introduced. The size of the polymeric matrix further is selected according to the method of delivery which is to be used. Preferably when an aerosol route is used the polymeric matrix and fugetactic agent are encompassed in a surfactant vehicle. The polymeric matrix composition can be selected to have both favorable degradation rates and also to be formed of a material which is bioadhesive, to further increase the effectiveness of transfer. The matrix composition also can be selected not to degrade, but rather, to release by diffusion over an extended period of time.
  • The delivery system may be a biocompatible microsphere that is suitable for local, site-specific delivery. Such microspheres are disclosed in Chickering et al., Biotech. And Bioeng., (1996) 52:96-101 and Mathiowitz et al., Nature, (1997) 386:.410-414.
  • Both non-biodegradable and biodegradable polymeric matrices can be used to deliver the fugetactic agents of the invention to the subject. Biodegradable matrices are preferred. Such polymers may be natural or synthetic polymers. Synthetic polymers are preferred. The polymer is selected based on the period of time over which release is desired, generally in the order of a few hours to a year or longer. Typically, release over a period ranging from between a few hours and three to twelve months is most desirable. The polymer optionally is in the form of a hydrogel that can absorb up to about 90% of its weight in water and further, optionally is cross-linked with multi-valent ions or other polymers.
  • In general, fugetactic agents are delivered using a bioerodible implant by way of diffusion, or more preferably, by degradation of the polymeric matrix. Exemplary synthetic polymers which can be used to form the biodegradable delivery system include: polyamides, polycarbonates, polyalkylenes, polyalkylene glycols, polyalkylene oxides, polyalkylene terepthalates, polyvinyl alcohols, polyvinyl ethers, polyvinyl esters, poly-vinyl halides, polyvinylpyrrolidone, polyglycolides, polysiloxanes, polyurethanes and co-polymers thereof, alkyl cellulose, hydroxyalkyl celluloses, cellulose ethers, cellulose esters, nitro celluloses, polymers of acrylic and methacrylic esters, methyl cellulose, ethyl cellulose, hydroxypropyl cellulose, hydroxy-propyl methyl cellulose, hydroxybutyl methyl cellulose, cellulose acetate, cellulose propionate, cellulose acetate butyrate, cellulose acetate phthalate, carboxylethyl cellulose, cellulose triacetate, cellulose sulphate sodium salt, poly(methyl methacrylate), poly(ethyl methacrylate), poly(butylmethacrylate), poly(isobutyl methacrylate), poly(hexylmethacrylate), poly(isodecyl methacrylate), poly(lauryl methacrylate), poly(phenyl methacrylate), poly(methyl acrylate), poly(isopropyl acrylate), poly(isobutyl acrylate), poly(octadecyl acrylate), polyethylene, polypropylene, poly(ethylene glycol), poly(ethylene oxide), poly(ethylene terephthalate), poly(vinyl alcohols), polyvinyl acetate, poly vinyl chloride, polystyrene, polyvinylpyrrolidone, and polymers of lactic acid and glycolic acid, polyanhydrides, poly(ortho)esters, poly(butic acid), poly(valeric acid), and poly(lactide-cocaprolactone), and natural polymers such as alginate and other polysaccharides including dextran and cellulose, collagen, chemical derivatives thereof (substitutions, additions of chemical groups, for example, alkyl, alkylene, hydroxylations, oxidations, and other modifications routinely made by those skilled in the art), albumin and other hydrophilic proteins, zein and other prolamines and hydrophobic proteins, copolymers and mixtures thereof. In general, these materials degrade either by enzymatic hydrolysis or exposure to water in vivo, by surface or bulk erosion.
  • Examples of non-biodegradable polymers include ethylene vinyl acetate, poly(meth)acrylic acid, polyamides, copolymers and mixtures thereof.
  • Bioadhesive polymers of particular interest include bioerodible hydrogels described by H.S. Sawhney, C.P. Pathak and J.A. Hubell in Macromolecules, (1993) 26:581-587, the teachings of which are incorporated herein, polyhyaluronic acids, casein, gelatin, glutin, polyanhydrides, polyacrylic acid, alginate, chitosan, poly(methyl methacrylates), poly(ethyl methacrylates), poly(butylmethacrylate), poly(isobutyl methacrylate), poly(hexylmethacrylate), poly(isodecyl methacrylate), poly(lauryl methacrylate), poly(phenyl methacrylate), poly(methyl acrylate), poly(isopropyl acrylate), poly(isobutyl acrylate), and poly(octadecyl acrylate).
  • There is also described herein pump-based hardware delivery systems, some of which are adapted for implantation. Such implantable pumps include controlled-release microchips. A preferred controlled-release microchip is described in Santini, JT Jr., et al., Nature, 1999, 397:335-338.
  • Use of a long-term sustained release implant may be particularly suitable for treatment of chronic conditions. Long-term release, are used herein, means that the implant is constructed and arranged to delivery therapeutic levels of the active ingredient for at least 30 days, and preferably 60 days. Long-term sustained release implants are well-known to those of ordinary skill in the art and include some of the release systems described above.
  • The isolated fugetactic agents described herein may be directly to the site at which there is inflammation, e.g., the joints in the case of a subject with rheumatoid arthritis, the blood vessels of an atherosclerotic organ, etc. For example, this can be accomplished by attaching an isolated fugetactic molecule (nucleic acid or polypeptide) to the surface of a balloon catheter; inserting the catheter into the subject until the balloon portion is located at the site of inflammation, e.g. an atherosclerotic vessel, and inflating the balloon to contact the balloon surface with the vessel wall at the site of the occlusion. In this manner, the compositions can be targeted locally to particular inflammatory sites to modulate immune cell migration to these sites. In another example the local administration involves an implantable pump to the site in need of such treatment. Preferred pumps are as described above. In a further example, when the treatment of an abscess is involved, the fugetactic agent may be delivered topically, e.g., in an ointment/dermal formulation. Optionally, the fugetactic molecules of the invention are delivered in combination with a non-fugetactic molecule (e.g., antiinflammatory, immunosuppressant, etc.).
  • Preferably the isolated fugetactic agents are administered to a subject in combination with a balloon angioplasty procedure. A balloon angioplasty procedure involves inserting a catheter having a deflated balloon into an artery. The deflated balloon is positioned in proximity to the atherosclerotic plaque and the site of inflammation, and is inflated such that the plaque is compressed against the arterial wall. As a result, the layer of endothelial cells on the surface of the artery is disrupted, thereby exposing the underlying vascular smooth muscle cells. The isolated fugetactic molecule is attached to the balloon angioplasty catheter in a manner which permits release of the isolated fugetactic molecule at the site of the atherosclerotic plaque and the site of inflammation. The isolated fugetactic molecule may be attached to the balloon angioplasty catheter in accordance with standard procedures known in the art. For example, the isolated fugetactic molecule may be stored in a compartment of the balloon angioplasty catheter until the balloon is inflated, at which point it is released into the local environment. Alteratively, the isolated fugetactic molecule may be impregnated on the balloon surface, such that it contacts the cells of the arterial wall as the balloon is inflated. The fugetactic molecule also may be delivered in a perforated balloon catheter such as those disclosed in Flugelman, et al., Circulation, v. 85, p. 1110-1117 (1992). See, also, e.g., published PCT Patent Application WO 95/23161 , for an exemplary procedure for attaching a therapeutic protein to a balloon angioplasty catheter. This procedure can be modified using no more that routine experimentation to attach a therapeutic nucleic acid to the balloon angioplasty catheter.
  • The invention will be more fully understood by reference to the following examples. These examples, however, are merely intended to illustrate the embodiments of the invention and are not to be construed to limit the scope of the invention.
  • Examples Materials and Methods Adult human venous blood sampling and sorting of subpopulations of peripheral blood T-cells
  • Peripheral blood was obtained from 12 healthy donors (age 25- 40 years, median age 31). Peripheral blood mononuclear cells (PBMC) were obtained from peripheral blood using Ficoll-Hypaque. PBMC were stained with anti-CD4 (PE), anti-CD8 (PE), anti-CD45RA (FITC), anti-CD45RO (FITC) (Becton-Dickinson, San Jose, CA). The CD4+CD45RA+, CD4+CD45RO+ and CD8+CD45RA+ or CD8+CD45RO+ subpopulations of peripheral blood cells were sorted using a FACS vantage sorter (Becton-Dickinson, San Jose, CA). The purity of each T-cell subpopulation was determined to be greater than 99% by immunophenotyping. Chemokine receptor expression including mean fluorescent intensities for chemokine receptors were also measured and compared for each T-cell subpopulation by staining sorted cells with anti-CXCR4 (PE) (Pharmagen/Becton Dickinson, San Diego, CA) directed against the cellular receptor for SDF-1α. Sorted adult peripheral blood T-cell subpopulations were cultured overnight in serum- free Iscoves modified medium (GIBCO BRL, Gaithersburg, MD), prior to their use in chemotaxis assays.
  • Processing of human fetal thymi and fetal blood and the preparation of purified subpopulations of human fetal thymocytes and fetal blood T-cells.
  • Human fetal thymi and peripheral blood samples were obtained from 16 to 22 week abortuses according to Institutional Review Board approved guidelines. Thymi were physically disaggregated and mononuclear cell suspensions generated. Thymocytes were stained with CD8 (FITC), CD3 (PE) and CD4 (APC) and sorted into purified subpopulations of triple negative immature thymocytes (TN) (CD3-, CD4-, CD8-) and mature double positive (DP) (CD4+CD8+) and single positive (SP) thymocytes (CD3+CD4+ or CD3+CD8+) by cell sorting. Fetal blood mononuclear cells (FBMC) were prepared from fetal blood using Ficoll-Hypaque as above. FBMC were labeled with CD8 (FITC), CD3 (PE), CD4 (APC) and anti-CD45RA (FITC). Purified populations of CD4+CD45RA+ and CD8+CD45RA+ T-cells were sorted cultured overnight in serum free Iscoves modified medium prior to use in chemotaxis assays.
  • Transmigration assays: Use of SDF-1α.
  • Three chemotaxis assays were used in this study. First, quantitative chemotaxis transmigration assays were performed using a transwell system (Coming Inc. , NY) (6.5mm diameter, 5µm pore size, polycarbonate membrane) as previously described 13. Purified T-cell and thymocyte subpopulations (5 x 104 cells) were added to the upper chamber of each well in a total volume of 150 µl of IMDM alone. SDF-1α (PeproTech, Rocky Hill, NJ) was used at concentrations of 10 µg/ml, 1 µg/ml, 100 ng/ml, 10 ng/ml and 0 ng/ml in the lower, upper or both lower and upper chambers of the transwell to generate a checkerboard analysis matrix of positive, negative and absent gradients of SDF-1α respectively. Cells were pretreated with pertussis toxin (PTX) 100 ng/ml for 1 hour, genistein (1µg/ml for 20minutes), herbimycin (1µM for 20 minutes) or wortmannin (1µM for 20 minutes) (Sigma Chemical Company, St Louis, MO) in a tissue culture incubator at 37°C and 5% CO2 prior to their addition to the upper chambers of the transwells in selected experiments.
  • Transwells prepared in this way were incubated for 3 hours at 37°C, 5% CO2 incubator. Cells were harvested from the lower chamber after 3 hours and cell counts, immunophenotype and CXCR-4 chemokine receptor expression determined.
  • The second semi-quantitative transmigration assay involved the use of a tissue culture insert (6.5 mm diameter, 0.45 µm pore size, polycarbonate membrane) (Millipore, Bedford MA). Adult peripheral blood T-cell subpopulations (1 × 105 cells) were plated into the wells of a 12-well tissue-culture dish in serum-free Iscove's medium. The tissue-culture plate was then incubated at 37°C for 1 hour to allow the cells to settle evenly over the base of the well. 0.1 ml of IMDM containing SDF-1α at final concentrations of 10 µg/ml, 1 µg/ml, 100 ng/ml, 10 ng/ml or 0ng/ml was then dispensed into the tissue culture insert. The tissue culture insert was gently placed in the center of the well of the 12-well tissue-culture plate leaving a gap of 0.3 cm between the edge of the insert and the edge of the well of the tissue culture plate. The well with transwell insert was incubated for up to 18 hours at 37°C on the heated stage of a microscope and viewed by time lapse videomicroscopy recording at 120 frames per hour or by Polaroid still photography. Maintenance of a gradient was assessed by loading of methylene blue into the insert and observing a preserved gradient of color over the incubation period.
  • In the third semi-quantitative migration assay, T-cell subpopulations (1 x 105 cells) were mixed with 0.5 ml of 0.5% methylcellulose (Stem Cell Technologies, Vancouver, BC) in sterile medium and plated in a 24 well plate. 5µl of PBS or SDF-1α at concentrations of 10µg/ml or 100ng/ml was then dispensed gently into the methylcellulose at a set position on the edge of the well prior to incubation for up to 18 hours on a heated (37°C) microscope stage and viewed by time lapse videomicroscopy.
  • Transmigration assays: Use of Thymic Stromal-Cell Medium(TSCM)
  • The chemotaxis assays used in this study were as for the SDF-1α described above. Quantitative chemotaxis transmigration assays were performed also using a transwell system. Serum free human fetal thymic stroma conditioned medium was added to the top, bottom or top and bottom chambers of the transwell in order to assess the chemotactic activities within the conditioned medium at dilutions of 1:2, 1:10 and 1:15 in serum free IMDM. Heat-inactivated TSCM was also used in the chemotaxis assay.
  • Cell migration was allowed to occur for 3 hours at 37°C in an incubator (5% CO2). Cells were harvested from the lower chamber after 3 hours and accurate cell counts, immunophenotypes, and chemokine receptor expression were determined for the cells which had emigrated from the upper chamber to the lower chamber.
  • Transmigration assays: Use of SDF-1, recombinant HIV-1IIIB gp 120 and inhibitors.
  • Two chemotaxis assays were used in this study. First, quantitative chemotaxis transmigration assays were performed as for the SDF-1α described above (and Kim, et al., Blood, 1998, 91:4434). Purified T-cell and thymocyte subpopulations (5 x 104 cells) were added to the upper chamber of each well in a total volume of 150 µl of IMDM alone. SDF-1α (PeproTech, Rocky Hill, NJ) was used at concentrations of 10 µg/ml, 1 µg/ml, 100 ng/ml, 10 ng/ml and 0 ng/ml in the lower, upper or both lower and upper chambers of the transwell to generate a checkerboard analysis matrix of positive, negative and absent gradients of SDF-1, respectively. Recombinant endotoxin free HIV-1IIIB gp 120 (Intracel, Rockville, MD) was also utilized as a chemokinetic stimulus for peripheral blood T-cells at concentrations of 2 ng/ml, 20 ng/ml, 200 ng/ml and 2 µg/ml.
  • Cells were pretreated with anti-CXCR-4 (Pharmingen, San Diego, CA) at 20 µg/ml for 30 minutes at room temperature, pertussis toxin (PTX) (100 ng/ml for 1 hour at 37°C), genistein (1 µg/ml for 20 minutes at 37°C), herbimycin (1µM for 20 minutes 37°C), wortmannin (1 µM for 20 minutes at 37°C), 8-Br-cAMP (100 µM for 15 minutes at room temperature) or 8-Br-cGMP (100 µM for 15 minutes at room temperature) (Sigma Chemical Company, St Louis, MO) prior to their addition to the upper chambers of the transwells in selected experiments. Transwells prepared in this way were incubated for 3 hours at 37°C /5% CO2 incubator. Cells were harvested from the lower chamber after 3 hours and cell counts performed using a hemocytometer.
  • In the second semi-quantitative migration assay, T-cell subpopulations (1 x 105 cells) were mixed with 0.5 ml of 0.5% methylcellulose (Stem Cell Technologies, Vancouver, BC) in sterile medium and plated in a 24 well plate. 5µl of PBS or SDF-1α at concentrations of 10µg/ml or 100ng/ml, was then dispensed gently into 0.5 ml of methylcellulose at a set position on the edge of the well prior to incubation for up to 18 hours on a heated (37°C) microscope stage and the movement of cells viewed by time lapse videomicroscopy.
  • Modulation of T cell infiltration into the site of a secondary immune response by SDF-1 in vivo
  • C57/B6 mice (Jackson Laboratories, ME) were obtained and primed subcutaneously with 100µg chicken ovalbumin (Sigma) containing less than 0.01% LPS by the limulus assay and dissolved in 100µl of Complete Freund's Adjuvant (CFA) (Sigma). Three days ova-primed mice were challenged by intraperitoneal (IP) injection with 100µg of ovalbumin (Ova) dissolved in 250µl of ddH2O. A second 250 µl IP injection was given to these mice the following day which contained murine SDF-1α at 100ng/ml, 1µg/ml or 5µg/ml. These mice were then euthanized 24 hours following the second IP injection and peritoneal lavage with 5 mls of PBS performed with direct visualization of the peritoneal sac and its contents in order to avoid peripheral blood contamination. Peritoneal fluid cells harvested in this way contained <0.1% red blood cells. Three groups of control mice were used. In the first, C57/B6 mice were euthanized prior to Ova-priming and a peritoneal lavage performed. The second group of control mice were primed subcutaneously and challenged with Ova intraperitoneally as above and then euthanized 24 hours later. The third group were primed and challenged with Ova as above and after 24 hours injected intraperitoneally with 250µl of ddH2O alone. These mice were euthanized and a peritoneal lavage performed 24 hours later. Three independent experiments were performed with two or three mice in each experiment.
  • Cells harvested from the peritoneal fluid were counted with a hemocytometer and their viability determined by Trypan blue exclusion and the total viable nucleated cell count per ml of peritoneal fluid was determined. Peritoneal fluid harvested in this way was found to contain <0.1% red blood cells. Hematological smears of peritoneal fluid were also prepared and stained with Giemsa. Flow cytometry using a FacsCalibur flow cytometer (Becton Dickenson) was then performed on peritoneal fluid cells using two panels of anti-murine T-cell monocyte antibodies (anti CD3-PE, anti CD8-PERCP, anti CD4-APC and anti TCRγδ, anti-CD11b, anti TCRαβ) (Caltag) and isotype controls. The proportion of T-cells of each subpopulation (CD3+, CD3+CD4+, CD3+CD8+) was determined as a percentage of the total nucleated cell fraction in the peritoneal fluid.
  • Results
  • SDF-1α is a chemokine produced by thymic and bone marrow stroma 12-15 which we evaluated for chemokinetic effects on subpopulations of human T cells, initially within circulating adult T cells and then fetal thymocytes and circulating T cells from the same fetus. Adult peripheral blood T-cells were isolated to a purity of ≥ 99% by flow cytometry into subpopulations of CD4+CD45RA+ and CD8+CD45RA+ (representing naive cells) and CD4+CD45RO+ and CD8+CD45RO+ (representing memory cells). All populations were represented (15 - 31 % of total lymphoid cells) and demonstrated high percentages of cells positive for the SDF-1α receptor, CXCR-4.
  • Using a standard transwell transmigration assay, SDF-1α demonstrated chemokinetic and chemoattractant activities for each subpopulation in relative order of CD8+CD45RA+ > CD4+CD45RA+ > CD4+CD45RO+ > CD8+ CD45RO+. Peak chemoattractant activities were seen at a concentration of 100 ng/ml with 97% of CD8+CD45RA+, 85% of CD4+CD45RA+, 79% of CD4+CD45RO+ and 51% of CD8+CD45RO+ T-cells transmigrating in response to a positive gradient of SDF-1α.
  • Increasing the SDF-1α concentration to 1 µg/ml and 10 µg/ml diminished the proportion of cells responding toward a concentration gradient and initiated the movement of T-cells away from SDF-1α,_a phenomenon we termed fugetaxis. Maximal fugetaxis was noted at 1µg/ml for CD8+CD45RO+, CD8+CD45RA+ and CD4+CD45RO+ and at 10 µg/ml for CD4+CD45RA+ cells. The proportion of cells moving away from the SDF-1α at 10 µg/ml was in relative order CD8+CD45RO+ ∼ CD4+CD45RA+ > CD8+CD45RA+ > CD4+CD45RO+ and was significantly greater than chemokinesis for each subset of cells (p < 0.01; Student t Test) (Figure 1A). Movement of cells away from 10 µg/ml SDF-1α was consistently 2 to 10 times higher than chemokinesis and, for CD8+CD45RA+ and CD8+CD45RO+ cells, greater than chemotaxis. As the concentration of SDF-1α was increased from 100 ng/ml to 10 µg/ml, movement away from SDF-1α increased for all but CD4+CD45RO+ cells whereas chemotaxis toward SDF-1α declined in all cells by up to a factor of 27-fold (Figure 1B).
  • In order to further explore fugetaxis, a second cell migration assay was developed in which a tissue culture insert served as a reservoir of SDF-1α and was placed in the center of a tissue culture well containing dispersed cells in serum free medium. When the insert contained 10 µg/ml of SDF-1α, adult peripheral blood T-cell subpopulations in the tissue culture well were observed to migrate towards the source of SDF-1α and a second population to migrate away. Only movement toward the insert was seen when the insert contained SDF-1α at 100 ng/ml. The polarization of T-cell subpopulations both towards and away from SDF-1α was also not seen when serum free medium was applied to the insert or when high concentrations of SDF-1α existed in both the insert and surrounding medium in the tissue-culture well.
  • To further validate the process of concentration dependent directional movement, a third transmigration assay was used. T-cell subpopulations (1 x 105 cells) were plated in a semi-solid methylcellulose matrix, SDF-1α was loaded at a fixed position and cell trafficking was recorded by time lapse videomicroscopy. The movement of a gradient across the methylcellulose was documented in independent experiments by monitoring a dye front through the culture; a process not achieving equilibrium over the duration of our incubations. Consistent movement of peripheral T-cell subpopulations was seen away from high concentrations of SDF-1α and towards low concentrations SDF-1α.
  • To investigate whether the movement away from high concentrations may be relevant for egress of mature cells from sites of differentiation, we examined cells representing stages of T cell development from the thymus and peripheral blood from individual abortuses. Fetal blood CD4+CD45RA+ and CD8+CD45RA+ T-cell subpopulations showed levels of CXCR-4 expression comparable to the same immunophenotypic populations in adult peripheral blood. Triple negative (TN), double positive (DP) and single positive (SP) fetal thymocytes were less often CXCR-4 positive than either fetal or adult peripheral blood T-cells (p < 0.05, Paired t Test). Mean fluorescent intensities of CXCR-4 on expressing thymocytes were also lower than on blood T-cells (p < 0.01, Paired t Test). Transmigration analysis of TN, DP and SP subpopulations of fetal thymocytes and CD4+CD45RA+ and CD8+CD45RA+ fetal blood T-cells demonstrated minimal cell movement in response to SDF-1α except in the most immature TN subset. In no thymocyte subpopulation was fugetaxis noted above background chemokinesis levels in response to SDF-1α, even at 1 µg/ml and 10 µg/ml.
  • Both CD4+CD45RA+ and CD8+CD45RA+ fetal blood T-cells showed significantly higher levels of chemotaxis and fugetaxis from SDF-1α than DP and SP thymocytes derived from the same fetuses (p < 0.05; Paired t Test). As with adult peripheral blood T-cells, fetal blood T-cells demonstrated maximal chemotactic responses at concentrations of SDF-1α of 100 ng/ml whereas fugetaxis was seen at concentrations of SDF-1α between 1µg and 10 µg/ml. Thus fetal samples from the same individual demonstrated the acquisition of fugetaxic ability only when T cells had exited the thymus into the circulation. The ability to undergo fugetaxis had marked differentiation stage specificity which was consistent with what we observed in the in vitro T cell differentiation system in which cells moved to the tissue culture well margins when fully differentiated.
  • We next evaluated whether the processes of cell movement toward or away from a chemokine signal were similarly mediated. Pertussis toxin (PTX), which inhibits Gαi coupled signalling, inhibited all cell movement responses to SDF-1α at concentrations between 10 ng/ml to 10 µg/ml (Figure 2). However, kinase inhibitor pre-incubation yielded distinct inhibition profiles for fugetaxis compared with chemotaxis. This was most readily seen with concentrations of SDF-1α that induced peak chemotaxis (100 ng/ml) or peak fugetaxis (10 µg/ml). The tyrosine kinase inhibitors, genistein and herbimycin, inhibited chemotaxis, but had minimal effect on fugetaxis. The PI-3 kinase inhibitor, wortmannin, comparably inhibited fugetaxis, chemotaxis and chemokinesis. This differential sensitivity to kinase inhibitors indicated unique signaling pathways mediating fugetaxis compared with chemotaxis to the same ligand.
  • Effects of TSCM (Thymic Stroma Conditioned Medium) on T-cell Transmigration
  • TSCM also demonstrated clear chemokinetic and chemoattractant activities for each of the four subpopulations of T-cells (see Figure 3). The order of chemoattractant activity was CD8+CD45RO+>CD4+CD45RO+> CD4+CD45RA+ >CD8+ CD45RA+. The chemoattractant effect of TSCM could be diluted out and was inhibitable by PTX. TSCM had minimal chemokinetic activities on all the T-cell subpopulations studied. The movement of cells away from TSCM was also demonstrated in all the T-cell subpopulations studied. In CD8+CD45RO+ and CD4+CD45RO+ cell populations, this movement of cells away from TSCM was reduced by dilution of the TSCM dose. However, the TSCM was enhanced by dilution in the CD8+CD45RO+ T-cell population. SDF-1α and SDF-1β were not detectable by immunoassay at concentrations above 10ng/ml in TSCM. Moreover, when fractionated, different fractions of TSCM had different effects on CD4+RO+ T cell transmigration. Chemoattractant activity resided with fraction 4 (pH 5.2, 0.25M NaCl) of the eluate, while fugetactic activity was associated with fraction 10 (pH 6.3, 0.5M NaCl) of the eluate.
  • The foregoing data demonstrate that subpopulations of human T-cells undergo purposeful movement away from high concentrations of SDF-1α and TSCM. The local concentrations of chemokines in tissue may be difficult to assess given the high affinity for proteoglycan binding, but has been postulated in the micromolar range16 and some chemokine concentrations in biologic fluids have been calculated at > 1µg/ml. Our data suggest that SDF-1α and TSCM participate in the process of emigration of mature cells from the thymus and provide one explanation for the observation made 8 years ago that transgenic mice expressing pertussis toxin driven by the lck promoter, were capable of forming normal, fully differentiated T cells, but that these cells failed to make their exit from the thymus1.
  • Fugetaxis is CXCR-4 mediated
  • We next evaluated whether the processes of cell movement toward or away from a chempkine signal were mediated via similar signal transduction pathways. Pre-incubation of cells with an anti-CXCR-4 specific monoclonal antibody that inhibits SDF-1 signaling resulted in inhibition of both chemotaxis and fugetaxis of T-cell subpopulations in response to concentrations of SDF-1α varying between 10ng/ml and 1µg/ml. In addition, PTX, which inhibits Gαi signaling, inhibited T-cell movement toward and away from SDF-1α at concentrations between 10ng/ml and 10µg/ml. Pre-incubation of cells with kinase inhibitors, however, resulted in distinct inhibition profiles for fugetaxis compared with chemotaxis. This was most readily apparent with SDF-1α concentrations that induced peak chemotaxis (100ng/ml) or peak fugetaxis (1µg/ml or 1µg/ml).
  • Chemokines, including SDF-1, have been shown to activate multiple signal transduction pathways including phosphatidylinositol 3-kinase (PI-3) and tyrosine kinase (Ganju, et al., J. Biol. Chem., 1998, 36, 23169; Turner, et al., J. Immunol., 1995, 5:2437; Turner, et al., J. Biochem. Soc. Trans., 1977, 2:216S). We postulated that the signals for fugetaxis or chemotaxis may have differential sensitivity to PI-3 and tyrosine kinase inhibitors. The PI-3 kinase inhibitor, wortmannin, comparably inhibited fugetaxis, chemotaxis and chemokinesis induced by SDF-1. In marked contrast, the tyrosine kinase inhibitors genistein and herbimycin inhibited chemotaxis, but had minimal effect on fugetaxis. Cyclic nucleotides serve as signaling intermediates for multiple G-protein coupled receptors (Daaka, et al., Nature, 1997, 6655, p88; Selbie, et al., Trends Pharmacol Sci, 1998, 3:87-93) and have been shown to convert neuronal growth cone responses to netrin-1 from repulsion to attraction (Song H-J, et al., Science, 1998, 281:1515). We noted that the membrane-permeable cAMP agonist, 8-Br-cAMP inhibited fugetaxis but not chemotaxis, and that inhibition was concentration dependent. Taken together, these data suggest that fugetaxis and chemotaxis result from the differential activation of signal transduction pathways in response to different concentrations of the same ligand.
  • HIV-1IIIB gp120 is a bidirectional cue for T-cell migration in vitro
  • To further confirm the role of CXCR-4 in mediating the fugetaxis signal, we used purified endotoxin free HIV-1IIIB gp120 (also known as X4-HIV-1 gp120) on CD8+ CD45RA+ T-cells (a subpopulation of human resting peripheral blood T-cells). SDF-1 independent and CD4 independent, chemotaxis and fugetaxis were observed in a concentration dependent manner (Figure 4), however, the proportion of cells undergoing chemotaxis was substantially less than that seen with SDF-1. High concentrations of HIV-1IIIB gp120 (2µg/ml) caused maximal migration of T-cells away from the HIV protein. In contrast, peak concentrations of HIV-1 gp120 of about 200ng/ml caused T-cells to move towards the recombinant retroviral protein. Similar to SDF-1, the bidirectional movement of T-cells induced by HIV-1IIIB gp120 was inhibited by pertussis toxin and the PI-3 kinase inhibitor wortmannin. In addition, movement away from HIV-1IIIB gp120 was differentially sensitive to inhibition by the cAMP agonist, 8-Br-cAMP. Furthermore, HIV-1 specific CTL clones derived from HIV-infected patients were shown to exhibit a bidirectional response to X4-HIV-1 gp120.
  • We conclude, therefore, that CXCR-4 ligands that include, but are not limited to, SDF-1α and β, met-SDF-1β, HIV- 1IIIB gp120, small molecules T134 and MD3100, T22 [Tyr5,12,Lys7]-polyphemusin II, agonists for such molecules, and the like, have a concentration dependent bifunctional effect on the migration of subpopulations of human T-cells.
  • SDF-1 concentrations in vivo alter inflammatory infiltrates
  • To evaluate whether the phenomenon of T cell movement away from high concentrations of SDF-1 was relevant to in vivo immune physiology, we immunized and subsequently challenged C57BL/6 mice with ovalbumin. We selected intraperitoneal challenge due to our ability to recover cells efficiently in quantities sufficient for flow cytometric analysis. Twenty-four hours following the challenge dose of Ova we injected mice with equivolume amounts of PBS, or low or high concentrations of SDF-1. After an additional 24 hours, Ova-challenged animals that had either not been injected or injected with PBS or SDF-1 were sacrificed and cells at the site of challenge quantitated and immunophenotyped. No significant differences in T cell accumulation were noted between those animals that were either sham injected, injected with PBS, or injected with SDF-1 at low dose (100 ng/ml) and all demonstrated marked T cell infiltration above that of baseline or 24 hours post antigenic challenge. In marked contrast, T cell infiltration with high dose SDF-1 (5 µg/ml) was blunted significantly below that of controls (p = 0.02) demonstrating the ability of high concentrations of a chemokine to mitigate and perhaps, reverse the potential of T cell accumulation at sites of inflammation. Of note, the concentrations of SDF-1 used had no demonstrable toxicity on T cells of the four subsets used in the in vitro analyses and no evidence of apoptosis on murine PBMC or CD3+ cells. (See Figure 5).
  • Purification of Putative Fugetaxins Derived from Cancer Cell lines Preparation of HepG2 Conditioned Medium (HepG2CM)
  • The HepG2 cell line (derived from a human hepatocarcinoma -ATCC no:HB-8065, ATCC, Manassas, VA) was grown to confluency in MEM + 10% heat inactivated fetal calf serum + penicillin/streptomycin + L-glutamine. Confluent HepG2 cells were then transferred into serum free medium for 48 hours and the serum free conditioned medium (HepG2CM) was harvested from these cultures. HepG2CM was filtered through a 0.4µm filter prior to use in transmigration assays or in the Fugetaxin separation system detailed below.
  • Preparation of fractions of HepG2 CM containing a putative Fugetaxin
    1. 1. Heparin is a highly sulfated glycosaminoglycan with the ability to bind most glycosylated secreted protein specifically. 200ml of HepG2CM were loaded onto a 5-ml HiTrap heparin column (Pharmacia) equilibrated with 10mM Na phosphate, pH 7. The column was eluted with step increasing concentration of NaCl. Major fugetactic activity was detected in the 0.3M NaCl eluted fraction (active fraction).
    2. 2. A cation exchange column, HiTrap Sp Sepharose (Pharmacia), was used as a second step of purification. The active fraction from the Heparin column was dialysized against 50mM MES, pH6.4 and loaded onto a 1-ml HiTrap Sp column equilibrated with 50mM MES, pH6.4. The column was then eluted with a linear gradient of NaCl concentration.
    3. 3. MonoQ is an anion exchanger with unique hydrophilic polymer particles designed for fast and high-resolution separation of proteins. The active fraction eluted from the HiTrap Sp column in the previous step was then dialysized with 20mM Tris, pH8.0 and further fractionated using a MonoQ HR5/5 column (Pharmacia). The column was then eluted with a linear gradient of NaCl concentration. By comparing fugetactic activity profile (using a standard transmigration assay) and a silver-stained SDS-PAGE gel, a particular band at approximately 110 kDa (p110) in fraction#18 was found always to correlate with such fugetactic activity.
    4. 4. Microsequencing of p110: To identify p110 candidate, MALDI-TOF MS (matrix-assisted laser desorption/ionization and time-of flight mass spectrometer) of tryptic digestion products were performed. The resulted peptide masses were used to search protein and DNA sequence database with an enhanced version of the FRAGFIT program (Henzel WJ, 1993, pnas 90, 5011 and Arnott D, 1998, Electrophoresis 19, 968). Mass spectrometer results indicated that p110 is the protein previous identified as inter-α-trypsin inhibitor heavy-chain II precursor (ITI H2). ITI H2 is a member of inter-α-trypsin inhibitor (ITI) family, which consist of a group of plasma proteins synthesized in the liver. Human ITI H2 is a glycoprotein composed of 946 amino acids (including a signal peptide of 18 aa) (SEQ ID NO: 1). It can undergo N-terminus and C-terminus cleavage steps after its secretion into plasma, eventually resulting in a 648 aa mature protein. ITI proteins, which is built up from heavy (H1, H2, H3, H4) and light (bikunin) chains, and belongs to the super-family of Kunitz-type protease inhibitors. Bikunin with a double-headed kunitz-type protease inhibitor sequence is the reason for the serine protease inhibitor activity. However, the array of proteases that are inhibited by ITI family members in vitro are more efficiently inhibited by other protease inhibitors in plasma. Therefore, the biological significance of ITI family members as protease inhibitors is unknown (Salier JP, Trends Biochem. Sci., 1990, 15:435). It has been suggested that ITI family members are involved in extracellular matrix stabilization (Chen L. et al., J. Biol. Chem., 1994, 269:28282), tumor invasion (Kobayashi H. et al.. J. Biol. Chem., 1995, 270:8361), metastasis (Kobayashi H. et al., Int. J. Cancer, 1995, 63:455) and arthritis (Hutadilok N. et al., Ann. Rheum. Dis., 1988,47:377).
    Biological activity of Fraction 18 of HepG2CM in vitro and in vivo
  • Fraction 18 at a 1:10 dilution in serum free medium (original fraction volume between 100-200 µl), was consistently shown to contain maximal fugetactic activity for Jurkat T-cells as described above. We examined the biology of the fugetactic activity of Fraction 18 with in vitro and in vivo transmigration assay systems.
  • Inhibitor sensitivity of Fraction 18 in vitro
  • Jurkat T-cells were pretreated with anti-CXCR-4 (Pharmingen) (at 20 µg/ml for 30 minutes at room temperature), pertussis toxin (PTX) (100 ng/ml for 30 minutes at 37 °C), genistein (1 µg/ml for 20 minutes at 37 °C), herbimycin (1µM for 20 minutes 37 °C), wortmannin (1 µM for 20 minutes at 37 °C), 8-Br-cAMP (100 µM for 15 minutes at room temperature) or 8-Br-cGMP (100 µM for 15 minutes at room temperature) (Sigma Chemical Company, St Louis, MO), prior to their addition to the upper chambers of the transwells in selected experiments. Transwells prepared in this way were incubated for 3 hours at 37° C /5% CO2 incubator. Cells were harvested from the lower chamber after 3 hours and cell counts performed. The effect of inhibitors on transmigration of Jurkat cell migration was examined. CXCR-4, genistein and herbamycin inhbited fugetaxis induced by Fraction 18 to 40 - 60% of the control level. PTX inhbited fugetaxis in response to Fraction 18 to 30% of the control level. The cAMP agonist, 8-Br-cAMP, resulted in 95% inhbition of fugetaxis whereas 8-Br-cGMP did not significantly inhbit fugetaxis.
  • Activity of Fraction 18 in vivo
  • C57 BL/6J mice (Jackson Laboratories, Bar Harbor, ME) were primed subcutaneously with 100µg chicken ovalbumin (Ova) (Sigma) containing less than 0.01% LPS by the limulus assay and dissolved in 100µl of Complete Freund's Adjuvant (CFA) (Sigma). Three days later Ova-primed mice were challenged by intraperitoneal (IP) injection with 100µg of Ova dissolved in 250µl of ddH2O. A second 250µl IP injection was given to these mice the following day, which contained a 1: 10 dilution of HepG2 CM fraction 18, fraction 22 (without significant fugetactic activity in vitro) or column pass through (PT). These mice were euthanized 3 and 24 hours following the second IP injection and peritoneal lavage with 5 mls of PBS performed with direct visualization of the peritoneal sac and its contents in order to avoid peripheral blood contamination. Peritoneal fluid cells harvested in this way contained <0.1% red blood cells.
  • Three groups of control mice were used. In the first, C57 BL/6J mice were euthanized prior to Ova priming and a peritoneal lavage performed. The second group of control mice were primed subcutaneously and challenged with Ova intraperitoneally as above and then euthanized 3 and 24 hours later. The third group were primed and challenged with Ova as above and injected intraperitoneally with 250µl of ddH2O alone 24 hours later. These mice were euthanized and a peritoneal lavage performed after a further 3 and 24 hours. Three independent experiments were performed with two or three mice in each group in each experiment.
  • Cells harvested from the peritoneal fluid were counted with a hemocytometer and their viability determined by trypan blue exclusion and the total viable nucleated cell count per ml of peritoneal fluid was determined. Peritoneal fluid harvested in this way was found to contain <0.1% red blood cells. Hematological smears of peritoneal fluid were also prepared and stained with Giemsa. Flow cytometry using a FACSCalibur flow cytometer (Becton Dickinson) was then performed on peritoneal fluid cells using two panels of anti-murine T-cell and monocyte antibodies (anti CD3-PE, anti CD8-Biotin, anti CD4-APC and anti TCRγδ, anti-CD11b, anti TCR αβ-FITC) (Caltag Laboratories, San Francisco, CA) and isotype controls (Caltag). Second-step staining of biotin-conjugated antibodies was performed using streptavidin-PERCP (Becton Dickinson). The proportion of T-cells of each subpopulation (CD3+, CD3+CD4+, CD3+CD8+) was determined as a percentage of the total nucleated cell fraction in the peritoneal fluid.
  • Fraction 18 was shown to abrogate the T-cell infiltrate induced by IP challenge with Ova. Fraction 22 or pass through did not significantly effect T-cell emigration from the site of antigen challenge (Figure 6).
  • According to this aspect of the invention, other members of the inter-α-trypsin inhibitor heavy-chain II precursor (ITI H2) family of polypeptides are believed to have similar fugetactic activity, and are therefore useful according to the invention. Such members include the polypeptides having sequences with GenBank Acc. Nos.: IYHU2, NP_034712, NP_002207, Q61703, P97279, O02668, CAA72308 (Y11545), BAA13939 (D89286), S54354, CAA49842(X70392), AAA60558(M18193), CAA30160(X07173), 1409219A, and/or AAA59195(M33033).
  • Kaposi's Sarcoma Herpes Virus and Fugetaxis
  • The human Kaposi's sarcoma herpes virus (KSHV) causes a number of different neoplastic disorders in an immunocompromised host including Kaposi's Sarcoma. KSHV has been shown to encode a number of human chemokine homologues including νMIP I and νMIP-II which are homologous to the human chemokines MIP-1α and MIP-1β. We postulate that the viral chemokines exert a fugetactic response on human T-cells thereby allowing the tumor to escape immune control.
  • We examined the chemotactic and fugetactic responses of human T-cell subpopulations and KSHV specific CTL clones to the KSHV proteins νMIP-I and νMIP-II and serum free supernatants from two cell-lines well known in the art that are chronically infected by KSHV and derived from human tumors, lines BCBL-1 and VG-1. BCBL-1 is a B cell line derived from a body cavity-based B lymphoma that is latently infected with HHV-8/KSHV; no EBV DNA is present in this line (Renne, R., W. et al., Nat. Med., 1996, 2:342-346).
  • Results
  • Significant fugetactic responses were discovered for CD4RA and CD4RO cells to VG-1 supernatants at a 1:2 dilution in serum free medium, and for CD4RO and CD8RO T-cells to BCBL-1 supernatant at a 1: 2 dilution.
  • T-cell subsets also show a clear chemotactic response to MIP-1α at a concentration of 10ng/ml. Of note, T-cells demonstrated a clear fugetactic response to νMIP-I but not νMIP-II at a concentration of 100ng/ml.
  • Bibliography
    1. 1. Chaffin KE and Perlmutter RM. A pertussis toxin-sensitive process controls thymocyte emigration. Eur.J.Immunol. 21: 2565-2573 (1991)
    2. 2. Craddock CF, Nakamoto B, Andrews RG, Priestley GV and Papayannopoulou T Antibodies to VLA4 integrin mobilize long-term repopulating cells and augment cytokine-induced mobilization in primates and mice. )
    3. 3. Doetsch RN and Seymour WF. Negative chemotaxis in bacteria. Life Sciences 9:1029-1037 (1970)
    4. 4. Bailey GB, Leitch GJ and Day DB. Chemotaxis by entamoeba histolytica. J Protozool 32:341-346 (1985)
    5. 5. Tsang N, Mcnab R and Koshland DE Jr. Common mechanism for repellents and attractants in bacterial chemotaxis. Science 181:60-69 (1973)
    6. 6. Repaske D and Adler J. Change in intracellular pH of Escherischia coli mediates the chemotactic response to certain attractants and repellents. J Bacteriol 145:1196-1208 (1981)
    7. 7. Tisa LS and Adler J. Cytoplasmic free-Ca2+ level rises with repellents and falls with attractants in Escherischia coli chemotaxis. Proc Natl Aca Sci U.S.A. 92:10777-10781 (1995)
    8. 8. Taylor BL and Johnson MS. Rewiring a receptor: negative output from positive input. FEBS Lett 425:377-381 (1998)
    9. 9. Wells TN, Power CA and Proudfoot AE. Definition, function and pathophysiological significance of chemokine receptors. Trends Pharmacol Sci 19:376-380 (1998)
    10. 10. Luster AD. Chemokines - chemotactic cytokines that mediate inflammation. N Engl J Med 338:436-445 (1998)
    11. 11. Baggiolini M. Chemokines and leukocyte traffic. Nature 392:565-568 (1998)
    12. 12. Bleul CC, Fuhlbrigge RC, Casasnovas JM, Aiuiti A and Springer TA. A highly efficacious lymphocyte chemoattractant, stromal cell-derived factor 1 (SDF-1). J Exp Med 184: 1101-1109 (1996)
    13. 13. Kim CH, Pelus LM, White JR and Broxmeyer HE. Differential chemotactic behavior of developing T-cells in response to thymic chemokines. Blood. 91: 4434-4443 (1998)
    14. 14. Tashiro K, Tada H, Heilker R, Shirozu M, Nakano T and Honjo T. Signal sequence trap : cloning strategy for secreted proteins and
    15. 15. Shirozu M, Nakano T, Inazawa J, Tashiro K, Tado H, Shinohara T and Honjo T. Structure and chromosomal localization of the human stromal cell-derived factor 1 (SDF1) gene. Genomics 28:495-500 (1995)
    16. 16. Bacon KB, Premack BA, Gardner P and Schall TJ. Activation of dual T cell signaling pathways by the chemokine RANTES. Science, 269:1727-30 (1995)
    17. 17. Noble PB and Bentley KC. Locomotory characteristics of human lymphocytes undergoing negative chemotaxis to oral carcinomas. Exp Cell Res 133; 457-461 (1981)
    Detailed Description of the Drawings
    • Figure 1 . SDF-1α induces fugetaxis, chemotaxis and chemokinesis that varies with input chemokine concentration. (A) Fugetactic (solid bars), chemotactic (striped bars) and chemokinetic (stippled bars) transmigration responses are shown in response to 10 µg/ml of SDF-1α for each of the four adult peripheral blood T-cell subpopulations (CD8+CD45RO+, CD4+CD45RA+, CD8+CD45RO+ and CD8+CD45RA+). Each data point represents the mean and S.E.M. of three independent experiments. (B) Depicts the percent change in fugetaxis and chemotaxis seen for the four adult peripheral blood subpopulations migrating in response to concentrations of SDF-1α between 100 ng/ml and 10 µg/ml.
    • Figure 2 . Both fugetaxis and chemotaxis are inhibitable by pertussis toxin. Dose response curves to SDF-1α are shown for fugetaxis and chemotaxis and individually for each of two adult peripheral blood T-cell subpopulations (CD4+CD45RA+ and CD8+ CD45RA+) without (-x-) and with (--x--) pretreatment with PTX. Each data point represents the mean and S.E.M. of three independent experiments.
    • Figure 3 . Chemotactic responses of peripheral bloodT cell subpopulations to Thymic Stroma Cell Medium (TSCM) Chemotaxis of purified T-cell subpopulations was examined in the transmigration system described above. In order to test positive chemotaxis or chemoattraction to TSCM, TSCM was placed at varying concentrations in IMDM in the lower chamber alone. To test for fugetaxis, TSCM was placed in the upper chamber to test for chemokinesis, TSCM was placed in the upper and lower chambers. Chemokinesis was examined for cells treated with undiluted TSCM, TSCM diluted to 1:10, heat inactivated TSCM (preincubated at 65°C for 60 minutes and TSCM in the context of cells pretreated with PTX. Chemotactic responses are shown for each of the three types of cell movement and individually for each of the four T-cell subpopulations (CD8+CD45RO+, CD4+CD45RA+, CD8+CD45RO+ and CD8+CD45RA+).
    • Figure 4 : CD8+CD45RA+ T-cells fugetax in a CD4-independent, CXCR-4 dependent, concentration dependent manner in response to recombinant HIV-1IIIB gp120. The concentration dependent chemotactic (----Δ----), fugetactic (-x-) and chemokinetic responses (----□----) of CD8+CD45RA+ T-cells to HIV- line gp120 was studied (Figure 4A). Migratory responses of to CD8+CD45RA+ T-cells to HIV- 1IIIB gp120 are shown following pretreatment of with anti-CXCR-4 (speckled bars), PTX (striped bars) and incubation in medium alone (solid bars) in response to concentrations of 20 ng/ml and 200 ng/ml of the recombinant HIV-1 glycoprotein (Figure 4B). Of note, no fugetaxis of CD8+CD45RA+ peripheral blood T-cells was seen in response to the CCR-5 specific gp 120 derived from HIV-I cm 235 (National Institutes of Health AIDS Reagent and Reference Program). CD8+CD45RA+ peripheral blood T-cells were incubated with the membrane permeable cyclic nucleotide inhibitors 8-Br-cAMP (----Δ----), 8-Br-cGMP (----□----) or in medium alone (-x-) prior to there use in transmigration assays with varying concentrations of HIV- 1 gp120. The concentration dependent chemotactic and fugetactic responses of the T-cells are shown (Figure 4C). Each data point represents the mean and S.E.M. of three independent experiments.
    • Figure 7 : Transmigration of human T-cells to supernatants derived from cell lines which are chronically infected with KSHV ( Fig. 7A : BCBL-1, and Fig. 7B :VG-1). Fugetactic (black bars) and chemotactic (striped bars) migratory responses are shown. Significant fugetactic responses are shown for CD4RA and CD4RO cells to VG-1 supernatants at a 1:2 dilution in serum free medium and for CD4RO and CD8RO T-cells to BCBL-1 supernatant at a 1: 2 dilution.
    SEQUENCE LISTING
    • <110> The General Hospital Corporation Poznansky, Mark C. Luster, Andrew D. Scadden, David T.
    • <120> PURPOSEFUL MOVEMENT OF HUMAN MIGRATORY
      CELLS AWAY FROM AN AGENT SOURCE
    • <130> M0765/7025WO/ERG/KA
    • <150> U.S. 60/128,272
      <151> 1999-04-08
    • <150> U.S. 60/168,952
      <151> 1999-12-03
    • <160> 1
    • <170> FastSEQ for Windows Version 3.0
    • <210> 1
      <211> 946
      <212> PRT
      <213> Homo Sapiens
    • <400> 1
      Figure imgb0001
      Figure imgb0002
      Figure imgb0003

Claims (15)

  1. Use of a fugetactic agent in the manufacture of a medicament for treating inflammation, autoimmune disease, an abscess, a transplant, an implant, atherosclerosis, myocarditis, infertility, or a tumor, the medicament containing a fugetactic amount of said agent for repelling immune cells from a specific site in a subject when the medicament is locally administered to the specific site in the subject, wherein the fugetactic agent is an agent having migratory cell repellent activity on a cell in a fugetactic amount, and having chemoattractant activity on the same cell at lower concentrations, wherein the fugetactic agent is SDF-1α or HIV-IIIIBgp120.
  2. Use according to claim 1 wherein the specific site is a site of inflammation.
  3. Use according to claim 2 further comprising the use of a non-fugetactic agent in combination with the fugetactic agent.
  4. Use according to claim 3 wherein the non-fugetactic agent is an antiinflammatory agent.
  5. Use according to claim 1, wherein the autoimmune disease is chosen from the group consisting of rheumatoid arthritis, uveitis, insulin-dependent diabetes mellitus, haemolytic anaemias, rheumatic fever, Crohn's disease, Guillain-Barre syndrome, psoriasis, thyroiditis, Grave's disease, myasthenia gravis, glomerulonephritis, autoimmune hepatitis or systemic lupus erythematosus.
  6. Use of a fugetactic agent in the manufacture of a medicament containing a fugetactic amount of said agent for repelling immune cells away from a specific site in a subject, wherein the fugetactic agent is SDF-1α or HIV-IIIIBgp120.
  7. Use according to claim 6 wherein the specific site is a chosen from the group consisting of transplanted tissue, an implant, an inflammation, an area surrounding a tumour or a germ cell containing site.
  8. A method for distinguishing mature T cells from immature T cells in vitro, comprising:
    contacting a fugetactic agent with a mixed population of mature and immature T cells and measuring cell movement relative to the fugetactic agent, wherein movement of the cells away from the fugetactic agent is indicative of mature T cells; wherein the fugetactic agent is SDF-1α or HIV-IIIIBgp120.
  9. Use according to claim 1 wherein the specific site in the subject is an area surrounding a tumour site and wherein the medicament is locally administered to said area surrounding a tumour to inhibit endothelial cell migration to the area surrounding a tumour site in the subject.
  10. Use according to claim 9 wherein the area surrounding the tumour site is not immediate to the tumour.
  11. Use according to claim 1 for treating infertility and premature labour, including premature delivery and impending miscarriage wherein the medicament repels immune cells from a germ cell containing site in a subject.
  12. Use according to claim 11 wherein the administration is local to a germ cell-containing site.
  13. A medicament containing a fugetactic agent for use in treating inflammation, autoimmune disease, an abscess, a transplant, an implant, atherosclerosis, myocarditis, infertility, or a tumor, wherein the medicament contains a fugetactic amount of said agent for repelling immune cells from a specific site in a subject when the medicament is locally administered to the specific site in the subject, wherein the fugetactic agent is an agent having migratory cell repellent activity on a cell in a fugetactic amount, and having chemoattractant activity on the same cell at lower concentrations, wherein the fugetactic agent is SDF-1α or HIV-IIIIBgp120.
  14. Use of a fugetactic agent to coat a material surface of an implant wherein the fugetactic agent is present in amount effective to repel immune cells from the material surface, wherein the fugetactic agent is SDF-1α or HIV-IIIIBgp120.
  15. An implant suitable for implanting into a subject wherein the implant comprises a material surface coated with an amount of a fugetactic agent effective to repel immune calls from the material surface, wherein the fugetactic agent is SDF-1α or HIV-IIIIBgp120.
EP00922067.4A 1999-04-08 2000-04-07 Purposeful movement of human migratory cells away from an agent source Expired - Lifetime EP1169348B1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US12827299P 1999-04-08 1999-04-08
US128272P 1999-04-08
US16895299P 1999-12-03 1999-12-03
US168952P 1999-12-03
PCT/US2000/009678 WO2000059941A1 (en) 1999-04-08 2000-04-07 Purposeful movement of human migratory cells away from an agent source

Publications (2)

Publication Number Publication Date
EP1169348A1 EP1169348A1 (en) 2002-01-09
EP1169348B1 true EP1169348B1 (en) 2016-02-24

Family

ID=26826444

Family Applications (1)

Application Number Title Priority Date Filing Date
EP00922067.4A Expired - Lifetime EP1169348B1 (en) 1999-04-08 2000-04-07 Purposeful movement of human migratory cells away from an agent source

Country Status (6)

Country Link
US (3) US6448054B1 (en)
EP (1) EP1169348B1 (en)
JP (1) JP2003502282A (en)
AU (1) AU4230900A (en)
CA (1) CA2367173A1 (en)
WO (1) WO2000059941A1 (en)

Families Citing this family (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6746482B2 (en) * 1994-10-17 2004-06-08 Baxter International Inc. Method for producing medical devices and devices so produced
US6448054B1 (en) 1999-04-08 2002-09-10 The General Hospital Corporation Purposeful movement of human migratory cells away from an agent source
US20020151515A1 (en) * 1999-06-18 2002-10-17 Roberts Bruce L. Preparation and use of superior vaccines
ES2231275T3 (en) 1999-10-08 2005-05-16 Affinium Pharmaceuticals, Inc. FAB INHIBITORS I.
WO2002036070A2 (en) * 2000-10-30 2002-05-10 Wayne State University Novel fish chemokine and methods for use
US6709655B2 (en) 2001-02-28 2004-03-23 Instituto Bioclon, S.A. De C.V. Pharmaceutical composition of F(ab1)2 antibody fragments and a process for the preparation thereof
DE60230934D1 (en) * 2001-04-06 2009-03-05 Affinium Pharm Inc FAB I INHIBITORS
DK1417488T3 (en) * 2001-06-07 2007-03-05 Chemocentryx Inc Cell migration assay
US7468253B2 (en) * 2001-06-07 2008-12-23 Chemocentryx, Inc. Method for multiple chemokine receptor screening for antagonists using RAM assay
DE10133870A1 (en) * 2001-07-12 2003-02-06 Chris P Lohmann Ophthalmic agent, use of EGF for the treatment of dry eye syndrome and insert for the administration of EGF to the eye
US20030065382A1 (en) * 2001-10-02 2003-04-03 Fischell Robert E. Means and method for the treatment of coronary artery obstructions
US20030100009A1 (en) * 2001-11-23 2003-05-29 George Jackowski Inter alpha trypsin inhibitor biopolymer markers indicative of insulin resistance
US7413866B2 (en) * 2001-11-30 2008-08-19 Chemocentryx, Inc. Compositions and methods for detecting and treating diseases and conditions related to chemokine receptors
US7871619B2 (en) * 2001-11-30 2011-01-18 Chemocentryx, Inc. Compositions and methods for detecting and treating diseases and conditions related to chemokine receptors
US7253007B2 (en) * 2001-11-30 2007-08-07 Chemocentryx, Inc. Compositions and methods for detecting and treating diseases and conditions related to chemokine receptors
US7442512B2 (en) * 2001-11-30 2008-10-28 Chemocentryx, Inc. Compositions and methods for detecting and treating diseases and conditions related to chemokine receptors
US20030194421A1 (en) * 2001-12-28 2003-10-16 Angiotech Pharmaceuticals, Inc. Treatment of uveitis
US20050271639A1 (en) * 2002-08-22 2005-12-08 Penn Marc S Genetically engineered cells for therapeutic applications
US7335759B2 (en) * 2002-12-02 2008-02-26 Universidad Nacional Autónoma de Méxica (UNAM) Recombinant immunogens for the generation of antivenoms to the venom of scorpions of the genus Centruroides
ES2518316T3 (en) 2002-12-06 2014-11-05 Debiopharm International Sa Heterocyclic compounds, their manufacturing methods and their use in therapy
EP1573066A1 (en) * 2002-12-06 2005-09-14 The General Hospital Corporation Methods and compositions relating to gradient exposed cells
WO2004058705A2 (en) * 2002-12-20 2004-07-15 Chemocentryx Inhibitors of the binding of chemokines i-tac or sdf-1 to the ccxckr2 receptor
WO2004072296A2 (en) * 2003-01-30 2004-08-26 The General Hospital Corporation Compositions for modulating immune cell activity and methods for detection thereof
DE602004016831D1 (en) * 2003-03-17 2008-11-13 Affinium Pharm Inc PHARMACEUTICAL COMPOSITIONS INHIBITORS OF FAB I AND OTHER ANTIBIOTICS CONTAINED
ES2339236T3 (en) * 2003-07-25 2010-05-18 Laboratorios Silanes, S.A. De C.V. ADMINISTRATION OF ANTIBODY FRAGMENTS F (AB ') 2 ANTI-TNF-ALFA.
US20060025335A1 (en) * 2004-01-30 2006-02-02 Kinane Thomas B Netrin compositions and methods of using the same
PL1828167T3 (en) 2004-06-04 2015-02-27 Debiopharm Int Sa Acrylamide derivatives as antibiotic agents
JP2008506703A (en) 2004-07-14 2008-03-06 ユニバーシティ オブ ユタ リサーチ ファウンデーション Netrin-related compounds and uses
US7351423B2 (en) 2004-09-01 2008-04-01 Depuy Spine, Inc. Musculo-skeletal implant having a bioactive gradient
US9789171B2 (en) * 2004-11-05 2017-10-17 The General Hospital Corporation Anti-fugetactic agents for the treatment of ovarian cancer
US7381802B2 (en) * 2005-04-15 2008-06-03 Universidad Nacional Autónoma De México (UNAM) Human antibodies that specifically recognize the toxin Cn2 from Centruroides noxius scorpion venom
PL1871804T3 (en) * 2005-04-21 2014-01-31 Chemocentryx Inc Antibodies that bind ccx-ckr2
KR20080075027A (en) * 2005-12-05 2008-08-13 아피늄 파마슈티컬스, 인크. Heterocyclylacrylamide compounds as fabi inhibitors and antibacterial agents
EP2687533B1 (en) 2006-07-20 2017-07-19 Debiopharm International SA Acrylamide derivatives as FAB I inhibitors
WO2008127355A2 (en) * 2006-10-06 2008-10-23 Celtaxsys, Inc. Chemorepulsion of cells
EP3255045A1 (en) 2007-02-16 2017-12-13 Debiopharm International SA Salts, prodrugs and polymorphs of fab i inhibitors
AU2008232739B2 (en) * 2007-03-30 2014-03-27 The Cleveland Clinic Foundation Method of treating ischemic disorders
MX2010001490A (en) * 2007-08-06 2010-03-04 Noxxon Pharma Ag Sdf-1 binding nucleic acids and the use thereof.
EP2234628B1 (en) 2007-12-14 2017-10-18 The Cleveland Clinic Foundation Compositions and methods of promoting wound healing
US8715654B2 (en) 2008-10-02 2014-05-06 Celtaxsys, Inc. Methods of modulating the negative chemotaxis of immune cells
WO2010042788A1 (en) * 2008-10-10 2010-04-15 Celtaxsys, Inc. Method of inducing negative chemotaxis
US20120283315A1 (en) 2009-08-28 2012-11-08 Penn Marc S Sdf-1 delivery for treating ischemic tissue
US11273250B2 (en) 2010-08-04 2022-03-15 Georgia Tech Research Corporation Devices, systems, and methods for excavating cancer cells
WO2013190384A1 (en) 2012-06-19 2013-12-27 Affinium Pharmaceuticals, Inc. Prodrug derivatives of (e)-n-methyl-n-((3-methylbenzofuran-2-yl)methyl)-3-(7-oxo-5,6,7,8-tetrahydro-1,8-naphthyridin-3-yl)acrylamide
DK2978839T3 (en) * 2013-03-28 2019-08-12 Ge Healthcare Bio Sciences Ab METHOD OF CELL CULTIVATION
US9775816B2 (en) * 2013-11-07 2017-10-03 The General Hospital Corporation Eluting matrix and uses thereof
CA2983758A1 (en) 2015-04-25 2016-11-03 The General Hospital Corporation Anti-fugetactic agent and anti-cancer agent combination therapy and compositions for the treatment of cancer
US11890348B2 (en) 2015-09-18 2024-02-06 The General Hospital Corporation Localized delivery of anti-fugetactic agent for treatment of cancer
WO2017049237A1 (en) * 2015-09-18 2017-03-23 The General Hospital Corporation Dba Massachusetts General Hospital Personalized approach to dosage of anti-fugetactic agent for treatment of cancer
PL3419628T3 (en) 2016-02-26 2021-05-31 Debiopharm International Sa Medicament for treatment of diabetic foot infections
US11325951B2 (en) 2016-09-09 2022-05-10 The General Hospital Corporation HSP fusion protein with anti-chemorepellant agent for treatment of cancer
US11453706B2 (en) 2016-09-09 2022-09-27 The General Hospital Corporation HSP fusion protein with anti-chemorepellant agent for treatment of infectious disease
WO2018049120A1 (en) 2016-09-09 2018-03-15 The General Hospital Corporation Ex vivo antigen-presenting cells or activated cd-positive t cells for treatment of cancer
WO2018102685A1 (en) * 2016-12-02 2018-06-07 The General Hospital Corporation Compositions and methods for reducing transplant rejection
CA3109902A1 (en) 2017-10-03 2019-04-11 Wallkill BioPharma, Inc. Treating diabetes with genetically modified beta cells
US10493233B1 (en) 2018-06-05 2019-12-03 Duke University Bi-directional access to tumors

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2557452B1 (en) 1983-12-28 1986-08-14 Roussel Uclaf NOVEL COMPOSITIONS FOR SKIN CARE CONTAINING PRIMER OIL AND RATE TISSUE TRAITS
CA1282727C (en) * 1985-07-11 1991-04-09 Michael R. Buchanan Chemorepellant compound
US5510418A (en) * 1988-11-21 1996-04-23 Collagen Corporation Glycosaminoglycan-synthetic polymer conjugates
US6399569B1 (en) * 1991-03-11 2002-06-04 Curis, Inc. Morphogen treatments for limiting proliferation of epithelial cells
US20020032313A1 (en) * 1991-03-29 2002-03-14 Genentech, Inc. Vascular endothelial cell growth factor antagonists
US5573934A (en) * 1992-04-20 1996-11-12 Board Of Regents, The University Of Texas System Gels for encapsulation of biological materials
US5514555A (en) 1993-03-12 1996-05-07 Center For Blood Research, Inc. Assays and therapeutic methods based on lymphocyte chemoattractants
FR2706771A1 (en) 1993-06-21 1994-12-30 Pelletier Jacques Formula for the treatment of certain cancers.
DE69434878T3 (en) * 1993-09-14 2014-07-17 Imperial Innovations Ltd. EOTAXIN = EOSINOPHIL CHEMOTOXIC CYTOKIN
JP3367581B2 (en) 1993-10-14 2003-01-14 小野薬品工業株式会社 Novel polypeptide, method for producing the same, DNA encoding the polypeptide, vector comprising the DNA, and host cells transformed with the vector
US5621671A (en) * 1995-03-17 1997-04-15 The United States Of America As Represented By The Secretary Of The Navy Digital simulation of organismal growth
US5624957A (en) * 1995-06-06 1997-04-29 Bristol-Myers Squibb Company Rary-specific retinobenzoic acid derivatives
WO1998009642A2 (en) * 1996-09-06 1998-03-12 The United States Of America, Represented By The Secretary, Department Of Health And Human Services Therapeutic chemokine antagonists
US5919776A (en) * 1996-12-20 1999-07-06 Merck & Co., Inc. Substituted aminoquinolines as modulators of chemokine receptor activity
IL132120A0 (en) * 1997-04-03 2001-03-19 Guilford Pharm Inc Biodegradable terephthalate polyester-poly (phosphate) polymers compositions articles and methods for making and using the same
US5994298A (en) * 1997-12-18 1999-11-30 Tsai; David Proteins for cancer cell specific induction of apoptosis and method for isolation thereof
WO2000007007A1 (en) * 1998-07-28 2000-02-10 Biometric Imaging, Inc. Device and method for cell motility assay
US6262228B1 (en) 1998-08-17 2001-07-17 Tularik Inc. IRAK3 polypeptides and methods
US6448054B1 (en) * 1999-04-08 2002-09-10 The General Hospital Corporation Purposeful movement of human migratory cells away from an agent source
US20020131953A1 (en) * 2001-03-14 2002-09-19 Ut Southwestern Medical Center In situ langerhans cell vaccine

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
D. M. BRAINARD ET AL: "Migration of Antigen-Specific T Cells Away from CXCR4-Binding Human Immunodeficiency Virus Type 1 gp120", JOURNAL OF VIROLOGY, vol. 78, no. 10, 15 May 2004 (2004-05-15), pages 5184 - 5193, XP055167887, ISSN: 0022-538X, DOI: 10.1128/JVI.78.10.5184-5193.2004 *
F. VIANELLO ET AL: "A CXCR4-Dependent Chemorepellent Signal Contributes to the Emigration of Mature Single-Positive CD4 Cells from the Fetal Thymus", THE JOURNAL OF IMMUNOLOGY, vol. 175, no. 8, 15 October 2005 (2005-10-15), pages 5115 - 5125, XP055167898, ISSN: 0022-1767, DOI: 10.4049/jimmunol.175.8.5115 *
FABRIZIO VIANELLO ET AL: "Murine B16 Melanomas Expressing High Levels of the Chemokine Stromal-Derived Factor-1/CXCL12 Induce Tumor-Specific T Cell Chemorepulsion and Escape from Immune Control", THE JOURNAL OF IMMUNOLOGY, THE AMERICAN ASSOCIATION OF IMMUNOLOGISTS, US, vol. 176, no. 5, 1 March 2006 (2006-03-01), pages 2902 - 2914, XP008146900, ISSN: 0022-1767 *
L. H. BLOCK: "Human migration inhibitory factor: purification and immunochemical characterization", JOURNAL OF EXPERIMENTAL MEDICINE, vol. 147, no. 2, 1 May 1978 (1978-05-01), pages 541 - 553, XP055167917, ISSN: 0022-1007, DOI: 10.1084/jem.147.2.541 *
PAPETA NATALIA ET AL: "long term survival of transplanted allogeneic cells engineered to express a t cell chemorepellent", TRANSPLANTATION, vol. 83, no. 2, 27 January 2007 (2007-01-27), pages 174 - 183, DOI: DOI: 10.1097/01.tp.0000250658.00925.c8 *
R H WEISBART ET AL: "Neutrophil migration inhibition factor from T lymphocytes (NIF-T): selective removal of biologic activity by human peripheral blood neutrophils, myelocytic leukemia cells, and differentiated HL-60 cells", JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950), 1 January 1982 (1982-01-01), UNITED STATES, pages 457, XP055167920, Retrieved from the Internet <URL:http://www.ncbi.nlm.nih.gov/pubmed/6976379> *
THARP WILLIAM G ET AL: "Neutrophil chemorepulsion in defined interleukin-8 gradients in vitro and in vivo", JOURNAL OF LEUKOCYTE BIOLOGY, FEDERATION OF AMERICAN SOCIETIES FOR EXPERIMENTAL BIOLOGY, US, vol. 79, no. 3, 1 March 2006 (2006-03-01), pages 539 - 554, XP002606478, ISSN: 0741-5400 *

Also Published As

Publication number Publication date
US20030017141A1 (en) 2003-01-23
US7141363B2 (en) 2006-11-28
WO2000059941A9 (en) 2002-01-31
JP2003502282A (en) 2003-01-21
EP1169348A1 (en) 2002-01-09
US20070026007A1 (en) 2007-02-01
US6448054B1 (en) 2002-09-10
AU4230900A (en) 2000-10-23
CA2367173A1 (en) 2000-10-12
US7775469B2 (en) 2010-08-17
WO2000059941A1 (en) 2000-10-12

Similar Documents

Publication Publication Date Title
EP1169348B1 (en) Purposeful movement of human migratory cells away from an agent source
US20180022793A1 (en) Chemorepulsion of cells
US20160193312A1 (en) Antifugetactic agents for the treatment of liver cancers
WO2010040029A1 (en) Methods of modulating the negative chemotaxis of immune cells
Farges et al. Odontoblasts in the dental pulp immune response
US20070134657A1 (en) Methods and compositions relating to gradient exposed cells
US7745578B2 (en) Fugetactic proteins, compositions and methods of use
Calvet et al. Trypanosoma cruzi–cardiomyocyte interaction: role of fibronectin in the recognition process and extracellular matrix expression in vitro and in vivo
US20020132224A1 (en) CaR receptor as a mediator of migratory cell chemotaxis and/or chemokinesis
Rezzani et al. Mast cells and the inflammatory response to different implanted biomaterials
Kang et al. Roles of protein phosphatase 1 and 2A in an IL-6-mediated autocrine growth loop of human myeloma cells
WO2005012351A2 (en) Novel functions for decay accelerating factor (daf) in inflammation
MXPA05006086A (en) Methods and compositions relating to gradient exposed cells
Phan Functional characterisation of an osteoclast-derived osteoblastic factor (ODOF)
Goerdt et al. 1–14 Prélude: Macrophages in Inflammation/15–35 Macrophages in Anti-Inflammation: Molecular Mechanisms/36–46 Macrophages in Anti-Inflammation: Apoptosis/47–56 Macrophages in Anti-Inflammation: Induction of Tolerance

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20011108

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

AX Request for extension of the european patent

Free format text: AL;LT;LV;MK;RO;SI

17Q First examination report despatched

Effective date: 20050913

REG Reference to a national code

Ref country code: DE

Ref legal event code: R079

Ref document number: 60049211

Country of ref document: DE

Free format text: PREVIOUS MAIN CLASS: C07K0014520000

Ipc: C07K0016240000

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

RIC1 Information provided on ipc code assigned before grant

Ipc: A61P 29/00 20060101ALI20150206BHEP

Ipc: C07K 16/24 20060101AFI20150206BHEP

Ipc: A61K 39/395 20060101ALI20150206BHEP

Ipc: C12P 21/00 20060101ALI20150206BHEP

Ipc: A61K 38/19 20060101ALI20150206BHEP

INTG Intention to grant announced

Effective date: 20150313

RIN1 Information on inventor provided before grant (corrected)

Inventor name: SCADDEN, DAVID, T.

Inventor name: POZNANSKY, MARK, C.

Inventor name: LUSTER, ANDREW, D.

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20150807

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 776690

Country of ref document: AT

Kind code of ref document: T

Effective date: 20160315

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 17

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 60049211

Country of ref document: DE

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: NL

Ref legal event code: MP

Effective date: 20160224

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 776690

Country of ref document: AT

Kind code of ref document: T

Effective date: 20160224

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160525

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160224

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160224

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160224

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160224

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160430

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160624

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160224

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160224

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 60049211

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160224

Ref country code: LU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160407

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20161125

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 18

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 19

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160224

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160224

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IE

Payment date: 20190429

Year of fee payment: 20

Ref country code: DE

Payment date: 20190429

Year of fee payment: 20

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20190425

Year of fee payment: 20

Ref country code: SE

Payment date: 20190429

Year of fee payment: 20

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CH

Payment date: 20190501

Year of fee payment: 20

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20190429

Year of fee payment: 20

REG Reference to a national code

Ref country code: DE

Ref legal event code: R071

Ref document number: 60049211

Country of ref document: DE

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

REG Reference to a national code

Ref country code: GB

Ref legal event code: PE20

Expiry date: 20200406

REG Reference to a national code

Ref country code: IE

Ref legal event code: MK9A

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF EXPIRATION OF PROTECTION

Effective date: 20200407

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GB

Free format text: LAPSE BECAUSE OF EXPIRATION OF PROTECTION

Effective date: 20200406